Ovid: Table of Contents
Transcription
Ovid: Table of Contents
I ﺷــﺮﻛـﺖ ﺭﻫــﺮﻭﺍﻥ ﻃــﺐ ((ﺛﻤﻴﻦ ))ﺑﺎ ﻣﺴﺌﻮﻟﻴﺖ ﻣﺤﺪﻭﺩ Medical Journals Print & CD-Rom Full Text Archives of 250 Medical Journals of CD-Rom Printable , Color For More Info. Please Contact Us : Tel & Fax : 021- 6429069 - 6432484 – 6947341 Second Floor , NO.135,Keshavarz Blvd./ Tehran 14187 , Iran – P.O.Box : 14185 - 157 Volume 16(6) November 2004 1. Editorial introductions. [Editorial introductions] 2. Making sure the treatment of myositis does not get "lost in translation". Isenberg, David[Myositis and myopathies: EDITORAL OVERVIEW] pg. 665-667 3. Clinical assessment in adult onset idiopathic inflammatory myopathy. Sultan, S M[Myositis and myopathies] pg. 668-672 4. Clinical assessment in juvenile idiopathic inflammatory myopathies and the development of disease activity and damage tools. Pilkington, Clarissa[Myositis and myopathies] pg. 673-677 5. Use of imaging to assess patients with muscle disease. Scott, David L a,b; Kingsley, Gabrielle H a,c[Myositis and myopathies] pg. 678-683 6. Is it really myositis? A consideration of the differential diagnosis. Nirmalananthan, Niranjanan a; Holton, Janice L b; Hanna, Michael G a,c[Myositis and pg. 684-691 myopathies] 7. Myositis specific autoantibodies: changing insights in pathophysiology and clinical associations. Hengstman, Gerald J.D a; van Engelen, Baziel G.M a; van Venrooij, Walther J b[Myositis and myopathies] pg. 692-699 8. Myositis: an update on pathogenesis. Christopher-Stine, Lisa a; Plotz, Paul H b[Myositis and myopathies] pg. 700-706 9. Have recent immunogenetic investigations increased our understanding of disease mechanisms in the idiopathic inflammatory myopathies? Chinoy, Hector; Ollier, William E.R; Cooper, Robert G[Myositis and myopathies] pg. 707-713 10. Illness and art: the legacy of Paul Klee. Varga, John[Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing pg. 714-717 syndromes: EDITORIAL OVERVIEW] 11. Raynaud phenomenon and the vascular disease in scleroderma. Kahaleh, M Bashar[Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing pg. 718-722 syndromes] 12. Autoantibodies in systemic sclerosis and fibrosing syndromes: clinical indications and relevance. Cepeda, Eduardo J; Reveille, John D[Raynaud phenomenon, scleroderma, overlap syndromes, pg. 723-732 and other fibrosing syndromes] 13. Cellular origins of fibroblasts: possible implications for organ fibrosis in systemic sclerosis. Postlethwaite, Arnold E a,b; Shigemitsu, Hidenobu b,c; Kanangat, Siva a,c[Raynaud pg. 733-738 phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes] 14. Recent advances in fibroblast signaling and biology in scleroderma. Pannu, Jaspreet; Trojanowska, Maria[Raynaud phenomenon, scleroderma, overlap syndromes, pg. 739-745 and other fibrosing syndromes] 15. Animal models of systemic sclerosis: insights into systemic sclerosis pathogenesis and potential therapeutic approaches. Christner, Paul J; Jimenez, Sergio A[Raynaud phenomenon, scleroderma, overlap syndromes, pg. 746-752 and other fibrosing syndromes] 16. Erratum. [Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes] 17. Bibliography Current World Literature. [Current World Literature] 18. List of journals scanned. [List of journals scanned] 19. Cumulative index to authors for volume 16. [Cumulative index to authors for volume 16: PDF pg. 753 pg. 754-778 pg. i-ii Only] 20. Cumulative index to subjects for volume 16. [Cumulative index to subjects for volume 16: PDF pg. iii-xi Only] 21. Cumulative contents for volume 16. [Cumulative contents for volume 16: PDF Only] pg. I-VI Editorial introductions Current Opinion in Rheumatology was launched in 1989. It is one of a successful series of review journals whose unique format is designed to provide a systematic and critical assessment of the literature as presented in the many primary journals. The field of rheumatology is divided into 15 sections that are reviewed once a year. Each section is assigned a Section Editor, a leading authority in the area, who identifies the most important topics at that time. Here we are pleased to introduce the Journal’s Section Editors for this issue. Section Editors David Isenberg, MD, FRCP Dr. Isenberg trained at St. Bartholomew’s Hospital, graduating in 1973. He undertook a variety of training posts in various North London hospitals before becoming a Sir Jules Thorn research fellow at University College and the Middlesex Hospitals, based in part in Ivan Riott’s Immunology Department. Subsequently, he undertook a further period of research as a Stanley Thomas Johnston research fellow in the Department of Haematology and Oncology at the New England Medical Centre, Tufts University, Boston, Massachusetts. These experiences led to the development of one of his major research interests: the link between structure, function, and origin of autoantibodies. His research laboratory, established at University College London in 1984, has pursued these topics ever since. Another major research interest has been seeking to improve the means of assessing disease activity in patients with autoimmune rheumatic diseases, notably lupus, myositis, and Sjögren syndrome. He has been Chairman of the British Isles Lupus Assessment Group (BILAG) for the past ten years, headed the Systemic Lupus International Collaborating Clinics group for five years, and is currently President of the British Society of Rheumatology. John Varga, MD Dr. Varga was born in Budapest, Hungary. He received his undergraduate training at Columbia University in New York, McGill University in Montreal, and Glasgow University in Scotland. He received his medical degree at New York University, and completed a residency in international medicine at Rhode Island Hospital, Brown University in Providence, Rhode Island, followed by a rheumatology fellowship at Boston University, Boston, Massachusetts. Dr. Varga was a post-doctoral research fellow of the Arthritis Foundation in the laboratory of Sergio Jimenez at the University of Pennsylvania. He joined the faculty of Jefferson Medical College in Philadelphia, Pennsylvania, in 1987. In 1995, he was appointed Director of Rheumatology at the University of Illinois College of Medicine, and in 2004, he was named The Gallgher Professor of Medicine at Northwestern University Feinberg School of Medicine in Chicago. Dr. Varga is Chair of the Scientific and Medical Advisory Board of the Scleroderma Foundation, and of the Abbott Scholar Advisory Board. He has served on National Institutes of Health Study Section panels in 1998. He has published over 120 peer-reviews articles, along with 60 reviews and book chapters, and two books. Dr. Varga directs an NIH-supported scleroderma research program focusing on basic, translational, and clinical aspects of this disease. He has trained over 20 clinical and research fellows. EDITORAL OVERVIEW Making sure the treatment of myositis does not get “lost in translation” David Isenberg Centre for Rheumatology, University College London, The Middlesex Hospital, London, UK Correspondence to David Isenberg, Centre for Rheumatology, University College London, The Middlesex Hospital, 4th Floor, Arthur Stanley House, 40-50 Tottenham Street, London W1T 4NJ, UK E-mail: [email protected] Current Opinion in Rheumatology 2004, 16:665–667 Separate tools need to be developed to distinguish these two items. To complete the understanding of the totality of the effect of the disease upon a patient and, by implication, the effect of a drug upon disease outcome, the patients’ own assessment of their health status is required. © 2004 Lippincott Williams & Wilkins 1040–8711 At a recent meeting at the Royal College of Physicians in London attended by senior members of the Department of Health, senior academics, and representatives of several major pharmaceutical companies the question of the future of clinical research was considered. Amongst the questions posed at this symposium was the critical issue of what has changed to make major pharmaceutical companies more interested in treating patients with autoimmune and other related diseases. From the perspective of autoimmunity there seem to be three critical changes: (1) The recognition that even for somewhat uncommon diseases such as systemic lupus erythematosus and scleroderma the number of patients who suffer from them worldwide is substantial. The number of patients with systemic lupus erythematosus (SLE) in the United States of America alone has been estimated at close to 250,000 [1]. (2) The huge amount of basic research undertaken into the etiopathogenesis of autoimmune diseases in the past twenty years is now paying dividends. A number of key molecules thought to be critical to the development of these disorders have been identified and agents that block or interfere with their function in some way have become available. (3) The ‘tools’ to assess patients with autoimmune diseases have been developed and many have been validated and shown to be reliable. An essential element in devising these tools has been the recognition that the distinction must be made between disease activity (implying ongoing active inflammation) and damage (implying permanent change). The lupus community has led the way in developing the different type of tools (for review, see references [2,3]). For those interested in myositis it is essential that, even though this disease has a much lower prevalence compared with related conditions such as lupus, Sjögren’s and scleroderma, an opportunity is not lost to undertake adequately controlled, reasonably sized trials of the more recently developed therapeutic agents. These agents suppress key molecules, as likely to be important in the development of myositis, as in the development of other autoimmune rheumatic diseases. In this brief review I will highlight some of the important international collaborative efforts that have been undertaken in the past three years to agree assessment tools for use in clinical trials of patients with myositis and briefly discuss some of the agents that might warrant a close examination of their therapeutic potential. Disease assessment in patients with myositis Following initial discussions at the European League against Rheumatism (EULAR) conference in Glasgow in 1999 the International Myositis Assessment and Clinical Studies (IMAC) group has formed and has actively explored the development of valid sensitive, reliable, and practical outcome measures that will be of use in randomized control trials for patients with myositis. Two disease activity tools have been devised, namely, the myositis intention to treat index (MITAX) and the myositis disease activity assessment visual analogue scale (MYOACT). The MITAX index is, in essence, a modification of the British Isles Lupus Assessment Group (BILAG) tool for assessing the disease activity in patients with lupus [4]. It is based on the principle of the physician’s intention to treat. Disease activity is divided 665 666 Myositis and myopathies into constitutional, dermatological, gastrointestinal, pulmonary, cardiac, and muscle organs/systems. Individual clinical features or combinations of features that were deemed likely to lead to the prescription of large doses of corticosteroids and/or immunosuppressive drugs define a grade A—the most active score in any organ or system. Those patients with known disease activity that is evident but requires somewhat lower doses of immunosuppression and/or drugs such as antimalarials or topical steroids sets the criteria we used to define a B grade. C grade in each organ or system defines patients with mild persistent activity. The D grade implies that the organ or system was once active but is no longer active and the E grade indicates that the organ or system has never been active. In contrast, the MYOACT consists of a series of 10-cm visual analogue scales completed by the physician reviewing the patients. The scales have been modified from those proposed in the assessment of patients with vasculitis [5]. The myositis damage index (MDI) is a comprehensive tool aimed at assessing the extent and severity of damage developing in different organs and systems. The first part of the index simply aims to count the items of developing in different organs or systems. This portion is, in essence, a modification of the Systemic Lupus International Collaborating Clinics/American College of Rheumatology damage index [6]. The other part, the myositis damage score (MYODAM), consists of a series of 10-cm visual analogue scales that aim to quantitate the severity of damage in the same organs or systems. ing in patients with myositis was considered in detail [9•]. Although some more recently described general immunosuppressives such as tacrolimus [10] and mycophenolate [11] have been tried in a few patients with myositis there is increasing interest in attempts to block more specific groups of cells or individual cytokines. For example anti-T lymphocyte globulin (ATG) appeared to increase muscle strength in one small study [12] and there are ongoing attempts to utilize antibodies to CD20positive B cells ([13•] Sultan S and Edwards JCW, personal communication). The first attempts to block the overexpression of tumor necrosis factor-␣ (TNF-␣) in patients with myositis have also been described [14]. In summary, the Oscar-winning film Lost in Translation describes the attempts of an American actor to make sense of a job opportunity that presents itself in Japan. Distractions are placed in his way but somehow he overcomes them. By analogy, there is an international consensus amongst rheumatologists and neurologists that we must make sensible attempts to assess both the effects of myositis and explore new opportunities to treat it. Some distractions lie ahead. There is for example a clear difference of opinion amongst interested parties about the utility of the current classification system for myositis [9•] but these obstacles must be overcome so that we may successfully translate our better understanding of the etiopathogenesis of myositis into more successful treatment for this seriously disabling disease. References and recommended reading Although no major international attempt has been made to develop a disease-specific tool for patient perception, use of the Short-Form 36 (SF36) in a group of patients with myositis has been reported [7]. As this tool has been used in a variety of diseases and is readily available in different translations it seems probable that it will be widely used in many control trials of patients with myositis. Papers of particular interest, published within the annual period of review, have been highlighted as: To start the process of determining the validity and reliability of the MITAX and MYOACT disease activity tools and the MYODAM disease damage measure, two ‘real patient’ exercises involving adult patients with myositis have been undertaken [8•]. The intraclass correlation coefficient among the physicians and the interrater reliability as assessed by a variation in the physicians rating of patients was generally good for most aspects of these tools. More extensive assessments are currently being undertaken to confirm their validity and reliability. • Of special interest •• Of outstanding interest 1 Rus V, Hochberg MC: The epidemiology of lupus erythematosus. In Dubois’ Lupus Erythematosus, edn 6. Edited by Wallace DJ, Hahn BH. Philadelphia: Lippincott Williams and Wilkins; 2002: 65–83. 2 Isenberg DA, Ramsey-Goldman R: Assessing patients with lupus: towards a drug-responder index. Rheumatology 1999, 38:1045–1049. 3 Ramsey-Goldman R, Isenberg DA: Systemic lupus erythematosus measures. Arthritis Care Res 2003, 49:S225–S233. 4 Hay EM, Bacon PA, Gordon C, et al.: The BILAG index: a reliable and valid instrument for measuring clinical activity in systemic lupus erythematosus. Q J Med 1993, 86:447–458. 5 Whiting-O’Keefe QE, Stone J, Hellman DB: Validity of a vasculitic index for systemic narcotising vasculitis. Arthritis Rheum 1999, 42:2365–2371. 6 Gladman DD, Ginzler E, Goldsmith C, et al.: The development and initial validation of the Systemic Lupus International Collaborating Clinics/America College of Rheumatology damage index for systemic lupus erythematosus. Arthritis Rheum 1996, 39:363–369. 7 Sultan S, Ioannou Y, Moss K, Isenberg DA: Outcome in patients with inflammatory myositis: morbidity and mortality. Rheumatology 2002, 41:22–26. 8 • Isenberg DA, Allen L, Farewell V, et al.: International consensus outcome measures for patients with idiopathic inflammatory myopathies. Development and initial validation of myositis activity and damage indices in patients with adult onset disease. Rheumatology 2004, 43:49–54. New agents At a recent international workshop at the European Neuromuscular Center Clinical Trials Network the questions of trial design and agents that might be suitable for test- Editorial overview: Myositis and myopathies Isenberg 667 9 • Hoogendijk JE, Amato AA, Lecky BR, et al.: The 119th ENMC international workshop: trial design in adult idiopathic inflammatory myopathies, with the exception of inclusion body myositis. Neuromuscul Disord 2004, 14:337– 345. 10 Oddis CV, Sciurba FC, Abu Elmgad K, Starzl TE: Tacrolimus in refractory polymyositis with interstitial lung disease. Lancet 1999, 353:1762. 11 Tousche AK, Mever M: Mycophenolate mofetil for dermatomyositis. Dermatology 2001, 202:341–343. 12 Lindberg C, Trysberg E, Tarkowski A, Oldfars A: Anti-T lymphocyte globulin treatment in inclusion body myositis. A randomised pilot study. Neurology 2003, 61:260–262. 13 • Edwards JCW, Szcepanski L, Szechinski J, et al.: Efficacy of B cell targeted therapy with rituximab in patients with rheumatoid arthritis. N Engl J Med 2004, 350:24–533. 14 Hengstman GJ, van den Horagen FH, Barrera P, et al.: Successful treatment of dermatomyositis and polymyositis with anti-tumour necrosis factor alpha. Preliminary observations. Eur Neurol 2003, 50:10–15. Clinical assessment in adult onset idiopathic inflammatory myopathy S.M. Sultan Purpose of review Outcome measures have been developed and validated for many rheumatic diseases and used in clinical trials. The clinical assessment and measures of improvement in clinical trials of patients with idiopathic inflammatory myopathy (IIM) has varied to date and over the past few years an attempt has been made to reach a consensus in defining this improvement. Recent findings The IMACS group (International Myositis Outcome Assessment Collaborative Study) has proposed core set measures for disease activity and damage assessment in adults. Distinguishing between activity and damage can be difficult and measures to ascertain this are discussed. Summary Significant progress has been made in developing international consensus in the assessment of IIM. Further developments are underway to assess the reliability and validity of measures that should capture the multisystem nature of this disease. Keywords idiopathic inflammatory myopathy, muscle strength, health related quality of life Introduction The assessment of disease activity and damage are clearly fundamental for the care of patients with idiopathic inflammatory myopathy (IIM) to optimize therapy and long-term prognosis. It is apparent that a fine line exists between under-treating a patient, which may lead to an increase in disease activity, and over-treating a patient, with the risk of serious morbidity from inappropriate therapy. It is generally accepted that to capture the totality of the effect of a disease on a patient, three aspects of the disease must be assessed: (1) Disease activity (which is potentially reversible with treatment) (2) Damage (defined as irreversible changes in anatomy, physiology, or function) accumulated since the onset of disease, albeit from the disease itself, comorbid conditions, or as a result of therapy (3) The patient’s own perception of the disease, as this is frequently different from the physician’s perception of the disease. Curr Opin Rheumatol 16:668–672. © 2004 Lippincott Williams & Wilkins. However, until a few years ago there was no agreement as to what set of measures should be included in the assessment of outcome of therapeutic trials in patients with IIM. Centre for Rheumatology, The Middlesex Hospital, University College Hospital, London, UK The clinical assessment and measures of improvement in clinical trials of patients with IIM has varied to date and over the past few years an attempt has been made to reach a consensus in defining this improvement. Measures are needed that capture the multisystem nature of the disease in patients with IIM. The patients frequently have cardiac, respiratory, cutaneous, skeletal (joints), and gastrointestinal involvement. In fact disease activity and damage in extramuscular organs may not necessarily be synchronous with the muscle involvement. With the prospect of new therapies, there is an urgency to develop a consensus on the assessment of patients with IIM. Correspondence to S.M. Sultan, Centre for Rheumatology, The Middlesex Hospital, University College Hospital, Arthur Stanley House, 40-50 Tottenham Street, London W1T 4NJ, UK Tel: 02073809230; fax: 02073809278; e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:668–672 © 2004 Lippincott Williams & Wilkins 1040–8711 This review highlights recent advances in the clinical assessment of myositis disease activity and damage. Current measures used to assess disease activity and damage Muscle strength testing Current evaluation of the extent of muscle involvement is most often accomplished through manual muscle 668 Adult onset idiopathic inflammatory myopathy Sultan 669 strength testing (MMT) and serial muscle enzyme measurements. The MMT is based on the manual muscle strength testing (MMT), based on the Medical Research Council War Memorandum scale, and is widely used in routine clinical care and clinical trials. Both the 6 point (0–5 scale) and the 12 point (0–10 scale) have been used and been shown to be sensitive in detecting change in strength when the muscles are moderately weak. However, the MMT is insensitive with lesser degrees of muscle weakness when using the 0 to 5 scale [1]. The total MMT score, rather than the score of individual muscles, appears to be most accurate for sequential monitoring [2]. The grading system is subjective and to an extent varies with the strength of the examiner. Error may be introduced due to the comprehension of the subject, motivation, and difference in stature of the subject relative to the tester. The use of MMT has been comprehensively reviewed by Harris-Love in a Workshop Report [3••]. More objective and sensitive measures of muscle strength are available, such as the hand-held dynamometry and fixed dynamometry [2,4]. However, they are time consuming and there is a lack of correlation with MMT, and these measures are therefore limited to research protocols [5]. However, neither manual MMT nor dynometers distinguish between active disease and damage although a significant improvement with treatment would suggest that the weakness was due to disease activity. Muscle enzymes Serum creatine kinase (CK) levels often do not correlate particularly well with muscle strength or physical function [6] although on an individual level they may be helpful in monitoring disease activity. Also, they do not assess extramuscular involvement and may be normal in some adults despite active disease (more often dermatomyositis). Lactate dehydrogenase (LDH) and alanine transferase (ALT) may also be elevated and correlate with disease activity, but are less sensitive than CK [6]. Elevation of the MB fraction of CPK and cardiac troponin T may be seen in the presence of regenerating myoblasts or myocarditis [7]. Serum cardiac troponin I is cardiac specific and is not produced by regenerating myoblasts, and may be useful in detecting myocarditis [8]. Assessment of extramuscular disease Idiopathic inflammatory myopathies are systemic diseases and assessment of constitutional symptoms, cutaneous, articular, gastrointestinal, respiratory, and cardiac systems are needed. Organ-specific measures currently used are valuable in the assessment of an individual patient but have not been developed into quantitative measures that could be used in clinical trials. Evaluation of cutaneous features and the activity and chronicity of individual lesions as well as assessment of periungual abnormalities has been shown to be sensitive in serial assessment of disease activity in patients with JDM [9]. Pulmonary involvement from underlying interstitial lung disease may be due to active inflammation, which is potentially reversible with treatment or from fibrosis, which is irreversible. Early in the disease, lung function tests may show a restrictive defect with a decreased diffusing capacity and hypoxemia with exercise and this can be used to monitor progress. The rate of decline may give some indication of the activity of the disease. However, high resolution computed tomography (HRCT) scanning [10], bronchoalveolar lavage, and occasionally lung biopsy [11] may be required to differentiate between activity and damage. A ground glass appearance on HRCT scanning is suggestive of an alveolitis or consolidation as part of an organizing pneumonia in active disease whereas honeycombing or diffuse alveolar damage occurs in the chronic, irreversible phase [11]. Bronchoalveolar lavage (BAL) is more invasive and may demonstrate an above-normal percentage of activated cytotoxic T cells [12] with a decrease in the CD4:CD8 ratio in the BAL fluid. Muscle imaging Muscle biopsy Magnetic resonance imaging (MRI) can demonstrate areas of muscle inflammation, edema suggesting active disease, and areas of fibrosis and calcification (ie, damage) [13]. It can be repeated sequentially and can be useful for guiding areas from which to take biopsies. T1-weighted images show muscle atrophy and fatty infiltration and T2-weighted images, which show increased water content as increased intensity best demonstrate areas of active muscle inflammation. Gadolinium enhancement is not helpful. MRI may show increased intensity in active disease, even when enzymes and other tests are normal [14]. Conversely false negatives do occur, even when there is ongoing muscle inflammation documented by muscle biopsy [15]. This is certainly useful at the time of diagnosis, but it is occasionally helpful in assessing clinical activity late in the course of the disease to determine activity versus damage. The yield may be further increased if it is MRI guided or at the very least a clinically weak muscle is biopsied. Magnetic resonance spectroscopy (MRS) provides a view of muscle metabolism comparing the ratio of muscle phosphorus contained in phosphocreatine to the level of inorganic phosphorous. This ratio is decreased in abnormal muscle. MRS is very sensitive (while not very spe- 670 Myositis and myopathies cific) and has been used to detect muscle abnormalities in the amyopathic variant of DM [14]. Although this form of imaging is not widely used in routine clinical practice this is due to the limitation in availability and cost. It is likely that the use of imaging will become a routine part of the evaluation of disease activity and damage in patients with IIM both at the time of diagnosis and follow-up. Certainly at present, it is still useful in the assessment of patients deemed refractory to treatment, where the distinction between damage and activity would mean reduction or discontinuation of immunosuppressive therapy. Recent developments in defining and assessing disease activity With the hope of new therapies on the horizon (eg, antiTNF ␣ drugs), there is an urgent need for tools that are reliable and validated for the assessment of disease activity and damage for the use in clinical trials. In clinical trials to date, various combinations of measures of disease activity have been used; it is important that a consensus is achieved to allow comparison of clinical trial data. A recent effort of an international group of specialists with expertise in these disorders has allowed substantial progress to be made. The IMACS group (International Myositis Outcome Assessment Collaborative Study) has focused on the development and validation of several measures to capture the totality of the effect of a disease on an individual. Three important domains have been recommended: disease activity, damage, and health related quality of life. Assessment of disease activity The IMACS have proposed core set measures for disease activity assessment in adults (Table 1) [16]. When making these assessments the physician takes into account information obtained from history, examination, and laboratory examination to create an overall impresTable 1. Proposed preliminary core set measure for disease activity assessment [16] Domain Global activity Muscle strength Physical function Laboratory assessment Extraskeletal muscle disease Core set measure (as proposed by the IMACS group) Physician global disease activity assessed by Likert or VAS Parent/patient global disease activity assessed by Likert or VAS MMT by a 0–10 scale or expanded 0–5 scale to include Proximal, distal, and axial muscles Validated patient/parent questionnaire of ADL (HAQ/CHAQ) Validated observational tool of function, strength, and endurance (CMAS) At least two serum muscle enzyme activities: CK, aldolase, LD, AST, or ALT A validated approach that is comprehensive and assesses cutaneous, gastrointestinal, joint, cardiac, and pulmonary activity must be developed sion of disease activity. The amount of clinical improvement deemed clinically significant is summarized in Table 2 [3]. Global disease activity as measured by Likert or visual analogue scale In the workshop report [3••] the use of a visual analogue scale (VAS) has been reviewed and it is suggested that physician global assessments correlate best with extramuscular activity, muscle strength, and physical function and is sensitive to change. Global measures can discriminate between active disease and damage [17]. Manual muscle strength testing Manual muscle strength testing has been selected by IMACS as the preferred method to assess muscle strength as one of the core set measures of IIM disease activity and damage [3••]. The reasons are that it has been partially validated, it is easily accessible, its costs are low, and it is easy to use. A total MMT score of 24 proximal, distal, axial muscles demonstrated good interrater and intrarater reliability, moderate sensitivity to change. A subset of eight axial and proximal muscles has been widely used as a primary endpoint in several adult PM/DM trials [3••]. Assessment of physical function The impact of the disease on the patient physical function and ability to self-care also must be assessed. Selfreported questionnaires such as the Stanford Health Assessment Questionnaire (HAQ) (developed in 1980) have been used widely. Assessment of eight domains is made: dressing, arising, eating, walking, hygiene, reach, grip, and social activities. It also consists of pain assessment, global severity, fatigue, and sleep VAS. The Medical Outcomes Study 36-item Short Form (SF-36) has been proposed for use in adult patients with myositis as a generic QOL measure. We have previously shown that the SF-36 scores differed between patients and controls in all domains, although there was no difference in the scores of the domains in patients with active disease compared with patients who had inactive disease [18]. Laboratory assessment In the workshop report by the IMACS group [3••] measurement of two muscle enzymes (CK, LDH, AST, ALT, aldolase) has been suggested. Inconsistent correTable 2. Consensus on the minimum percentage change in the myositis core set measures to classify a patient as clinically improved [3••] Core set domain Global activity assessment Patient global activity assessment Muscle strength Physical function Muscle-associated enzymes Extramuscular activity assessment Adult specialists, Median % change (25th, 75th percentile) 20 (20, 25) 20 (20, 25) 15 (10, 20) 15 (10, 20) 30 (20, 50) 20 (20, 28) Adult onset idiopathic inflammatory myopathy Sultan 671 lation of CK with muscle histology or muscle activity is multifactorial and is in part due to suppression of CK by corticosteroid therapy loss of intrinsic CK activity in the presence of muscle atrophy. However, in an individual patient CPK remains a helpful guide in predicting clinical relapse. LDH may be the most clinically useful muscle enzyme in patients with longstanding disease [6]. patients with underlying OA. The low agreement in muscle assessment when using the MITAX appears to be difficulty in attributing weakness due to activity and that due to damage when scoring ‘loss of function’. This aspect will need further investigation and possibly modification of the assessment tool. The face validity of the tool is also being evaluated. Extraskeletal muscle disease Consensus approach to the assessment of disease damage Two comprehensive quantitative measures have been developed by the IMACS group to assess the disease activity in target organs other than muscle [19•]: The Myositis Intention to Treat Index (MITAX) and the Myositis Activity Assessment by Visual Analogue Scales (MYOACT). The MITAX is in fact based upon the BILAG lupus activity index and is an intention-to-treat index. It represents the physicians’ judgment of how active the patients’ disease has been in the previous 2 weeks. Seven target organs are assessed: constitutional, mucocutaneous, joints, gastrointestinal (GI), respiratory (RS), cardiovascular (CV), and muscle. Also a physician global VAS is obtained for the disease activity. Each clinical feature is scored as not present, improving, the same, worse, or new. Scores for each system range from category A score (implying very active disease with the requirement for high levels of steroids and/or immunosuppressive drugs), category B (active but controlled disease), category C (stable, relatively mild disease), category D (disease that was once active but is no longer so), and category E (organ never affected by the disease). In the first test of its performance (a ‘real patient’ exercise involving seven patients and seven physician assessors), the interrater reliability of the MITAX was shown to be good except for the skeletal and muscle assessments. The MYOACT was considered good for mucocutaneous, average for GI, CV, RS, and muscle [18]. A further interreliability exercise has been undertaken as part of a multicenter effort (unpublished data). Six centers have participated to date: University College London, St. Georges Hospital (London), Manchester, Pittsburgh, Sweden, and Prague. The activity indices were filled in both by the local physician and myself (SS) independently of one another. To date a total of 105 patients have been seen with the local physician. Correlation between physicians has been reported as Intraclass Correlation Coefficients (ICC). An ICC > 0.65 has been used as indicating a high level of agreement. Again there appears to be good agreement in the assessment of most systems. The agreement was low in the assessment of skeletal and muscle involvement when using the MITAX and low in constitutional, mucocutaneous, and gastrointestinal assessment for the MYOACT. In the assessment of the skeletal system disagreement occurred when the patient had overlap RA or To date there is limited information as how best to define or assess damage. MRI as discussed earlier certainly may have a greater role to play in the future in differentiating between inflammation, fibrosis, and atrophy. The IMACS group has suggested that further investigation is required as to what measures should be used in clinical trials [3••]. At present the following have been suggested: the physician global damage assessments (as measured by a VAS); the HAQ/CHAQ as measures of physical function—this has been shown to measure a cumulative decline in function in adult patients and the Myositis Damage Index (MDI) [3••]. The MDI has been devised to assess the extent and severity of damage developing in different organs and systems. The MDI is an assessment of accumulation of damage since the onset of the disease. No distinction is made as to whether this is as a result of the disease itself, comorbid conditions, or as a result of therapy as not infrequently it is difficult to differentiate between these. It has been agreed that for changes to constitute damage they must have been present for at least 6 months (or the pathology that lead to the feature must have been present for at least 6 months). A comprehensive organ-based assessment is made including: muscle, skeletal (joints and damage as a result of osteoporosis), mucocutaneous, pulmonary, cardiovascular, peripheral vascular, gastrointestinal, endocrine, ocular, and infection. Malignancy and death are also recorded. We have also assessed the interrater reliability of this tool (unpublished). The between rater reliability for the damage index was fair. In most systems, the ICC was > 0.7. The agreement in the cardiovascular, peripheral vascular disease, malignancy was relatively low. The agreement for the VAS for the damage index was generally good except for peripheral vascular disease. The agreement for the global VAS for the damage index was very good ICC > 0.8. Assessment of health related quality of life No disease outcome assessment would be complete without an assessment of the patients’ perception of their disease. Various health related quality of life (HRQOL) measures are available, but none have been validated in IIM. The Nottingham Health Profile [20] and the Short Form-36 [18] have been used in IIM. The 672 Myositis and myopathies IMACs group has suggested using the SF-36 as it has been extensively validated in other rheumatic diseases and its use has been widespread. 8 White GH, Tideman PA: Increased Troponin I in a patient with dermatomyositis. Clin Chem 2001, 47:1130–1131. 9 Pachman LM, Sundberg J, Maduzia , et al.: Sequential studies of nailfold capillaries vessels in 10 children with juvenile dermatomyositis: correlation with disease activity score but not von Willebrand factor antigen. Arthritis Rheum 1996, 38(suppl):360. 10 Akira M, Hara H, Sakatani M: Interstitial lung disease in association with polymyositis-dermatomyositis: long-term follow-up CT evaluation in seven patients. Radiology 1999, 210:333–338. 11 Tazelaar HD, Viggiano RW, Pickersgill J, et al.: Interstitial lung disease in polymyositis and dermatomyositis: clinical features and prognosis as correlated with histologic findings. Am Rev Respir Dis 1990, 141:727–733. 12 Kourakata H, Takada T, Suzuki E, et al.: Flow cytometric analysis of bronchoalvelar lavage fluid cells in polymyositis/dermatomyositis with interstitial pneumonia. Respirology 1999, 4:223–228. 13 Park JH, Olsen NJ, King L, et al.: MRI and P-31 magnetic resonance spectroscopy detect and quantify muscle dysfunction in the amyopathic and myopathic variants of dermatomyositis. Arthritis Rheum 1995, 38:68–77. 14 Pachman LM, Crawford S, Morrelo F, et al.: MRI directed muscle biopsy for assessment of juvenile dermatomyositis response to therapy: comparison on initial and follow up biopsies using histological rating scale evaluating disease severity/chronicity. Arthritis Rheum 1996, 38(suppl):360. Conclusion In conclusion, much progress has been made in developing a consensus as to the combination of assessments that would best define disease activity and damage in patients with IIM. Further studies are underway to assess the validity and reliability of these assessment tools. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest 1 Kendall FP, McCreary EK, Povance PG: In Muscles: Testing and Function, edn 4. Baltimore: Williams and Wilkins: 1993. 2 Hinder KA, Hinderer SR: Muscle strength development and assessment in children and adolescents. In Muscle Strength. Edited by Harms-Ringdahl K. Edinburgh: Churchill Livingstone; 1993: 93–140. 15 Dorph C, Nennesmo I, Lundberg I: Percutaneous conchotome muscle biopsy: a useful diagnostic and assessment tool. J Rheumatol 2001, 60:423– 426. 3 Rider LG, Giannini EH, Harris-Love M, et al.: Defining clinical improvement in adult and juvenile myositis. J Rheumatol 2003, 30:603–617. •• An excellent and comprehensive review of measures used to define disease activity and damage to date. It ties together the evidence for the current use of measures and efforts to standardize these outcome measures for use in clinical trial. 16 Miller FW, Rider LG, Chung YL, et al.: Proposed preliminary core set measures for disease outcome assessment in adult and juvenile idiopathic inflammatory myopathies. Rheumatol 2001, 40:1262–1273. 17 Rider LG: Outcome assessment in the adult and juvenile idiopathic inflammatory myopathies. Rheum Dis Clin North Am 2002, 28:935–977. Sultan SM, Ioannou Y, Moss K, Isenberg DA: Outcome in patients with idiopathic inflammatory myositis: morbidity and mortality. Rheumatology 2002, 41:22–26. 4 Escolar DM, Henricson EK, Mayhew J, et al.: Clinical evaluator reliability for quantitative and manual muscle testing measures of strength in children. Muscle Nerve 2001, 24:787–793. 18 5 Watkins MP, Harris BA: Evaluation of skeletal muscle performance. In Muscle Strength. Edited by Harms-Ringdahl K. Edinburgh: Churchill Livingstone; 1993: 19–36. 19 • 6 Rider LG, Miller FW: Laboratory evaluation of the inflammatory myopathies. Clin Diagn Lab Immunol 1995, 2:1–9. 7 Bodor GS, Survant L, Voss EM, et al.: Cardiac troponin T composition in normal and regenerating human skeletal muscle. Clin Chem 1997, 43:476– 484. Isenberg DA, Allen E, Farewell V, et al.: International consensus outcome measures for patients with idiopathic inflammatory myopathies. Development and initial validation of myositis activity and damage indices in patients with adult onset disease. Rheumatology 2004, 43:49–54. A remarkable international effort to standardize the reporting of outcome in IIM. The first workshop report assessing the reliability of the activity and damage index. 20 Chung YL, Houssien DA, Scott DL: Health status in inflammatory muscle disease. Arthritis Rheum 1997, 40(suppl):S115. Clinical assessment in juvenile idiopathic inflammatory myopathies and the development of disease activity and damage tools Clarissa Pilkington Purpose of review In the past few years, adult and pediatric experts in the field have combined their skills to achieve major advances in clinical research on pediatric inflammatory myopathies. As rare diseases, it has become recognized that international trials are needed to assess drug treatments and their long-term sequelae. Tools for assessing specific aspects of disease are available and have been studied to better understand their performance strengths and limitations. These tools have been incorporated into disease activity and damage scores that have been developed by international consensus and are now being evaluated. Recent findings This review focuses on the tools whose evaluations have been published in the past year and includes methods to assess muscle strength and function in children with myositis. Two international collaborative efforts have focused on disease activity and damage assessments, and their findings are discussed. Some papers highlighting further diagnostic investigations are also reviewed. Summary The standardization of assessment tools has enormous implications for future clinical practice: (1) Clinicians who are not experienced in these rare disorders will be able to evaluate their patients appropriately and highlight important clinical signs that are of prognostic importance. (2) International drug trials can be undertaken and thus improve our understanding of the impact of medication on the disease process. (3) Follow-up studies can be undertaken with sufficient numbers of patients to answer questions about the long-term sequelae of the disease and its treatments. Keywords inflammatory myopathies, juvenile dermatomyositis, disease activity, disease damage Curr Opin Rheumatol 16:673–677. © 2004 Lippincott Williams & Wilkins. Great Ormond Street and University College London Hospitals and Juvenile Dermatomyositis Research Centre, Institute of Child Health, London, United Kingdom The author thanks the Cathal Hayes Research Foundation for supporting her involvement in this international work. Correspondence to Clarissa Pilkington, MD, Institute of Child Health, Department of Rheumatology (JDRC), 30 Guilford Street, London WC1N 1EH, UK Tel: 0207905 2667; fax: 0207905 2672; e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:673–677 Abbreviations CHAQ CMAS JDM Childhood Health Assessment Questionnaire Childhood Myositis Assessment Scale juvenile dermatomyositis © 2004 Lippincott Williams & Wilkins 1040–8711 Introduction Juvenile dermatomyositis (JDM) is the most common of the juvenile-onset inflammatory myopathies. Even so, it is a rare disease with a reported incidence of approximately three cases per million children [1•,2]. Most general pediatricians in a district general hospital may only see two cases during their working lives, and many pediatric rheumatology centers will only see three or four new cases in a year. This makes it difficult for doctors to gain enough expertise through clinical experience of their own patients to further the understanding of this “orphan disease.” As such, it is impossible for research to move forward without collaboration. In the past few years, there has been an increasing collaborative effort between adult and pediatric rheumatologists, neurologists, and dermatologists to pool expertise and develop assessment tools. Over the past year, there have been several papers published as a result of these collaborations. Inflammatory myopathies in childhood include JDM overlap syndromes (such as JDM- scleroderma overlap) and the extremely rare polymyositis. The prognosis for JDM before the introduction of steroids was a 33% mortality rate, with 33% of the survivors being left with a severe disability [3]. There have been no double-blind trials on the use of corticosteroids, but the mortality rate has been reduced to less than 10% [4]. More aggressive treatment is associated with improved outcomes [5]. However, there are still patients who do not respond to first-line treatments and continue to have active disease. Newer treatments are available, but physicians need to be able to evaluate outcome data to select which treatments are most suitable for individual cases. To compare the effectiveness of drug treatments, there needs to be a standardized set of tools for measuring and assessing disease activity. The hope of all physicians is to control 673 674 Myositis and myopathies disease activity, reduce the length of the disease process, and prevent accumulation of damage from the disease or its treatments. Outcome studies are needed to allow physicians to evaluate the risks and benefits of differing treatments: these need standardized disease damage assessment indices to allow multicenter research. Another element in the process of researching rare diseases is the need to “collect” sufficient numbers for studying. Again, there has been an increase in the collaborative efforts of clinicians caring for these diseases, and registries have been set up. In the United States, there is the National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) Juvenile Dermatomyositis Research Registry; and in the United Kingdom and Ireland, there is the Juvenile Dermatomyositis National Registry and Repository. Incidence of juvenile dermatomyositis The reported incidence of JDM varies from 1.9 to 4.2 cases per million children [6,7]. These reports have been derived by extrapolating from the study of cases from referral centers, apart from the 1995 United Kingdom study, which was a nationwide study over the course of 1 year [7]. However, the incidence may vary from one year to another and between different racial groups. The National Institute of Arthritis and Musculoskeletal and Skin Diseases study [1•] used two registries in a two-source capture–recapture analysis to estimate as accurately as possible the annual incidence of JDM in the United States between 1995 and 1998. It also looked at the incidence within different racial groups. Their findings are reproduced in Table 1. This demonstrates that the incidence rate varies between years with an average of 3.2 per million children per year. Breakdown by ethnicity showed that Hispanics had a slightly lower incidence (2.7 per million) than white non-Hispanics (3.4 per million) and African-American non-Hispanics (3.3 per million). Therefore, future epidemiologic studies will need to take race and variations between years into account. Pediatric rheumatology assessment tools Classic JDM has a characteristic rash and symmetric proximal muscle weakness [8]. It may have an associated vasculitis that can affect multiple organs and result in skin ulceration and calcinosis. In the assessment of JDM, there needs to be assessments of multiple systems Table 1. Juvenile dermatomyositis (2–17 years of age) incidence estimates in the United States Year 1995 1996 1997 1998 Average Adapted from [1]. Incidence per million 95% CI 4.1 2.5 2.7 3.4 3.2 3.1–5.1 2.1–2.9 2.5–3.0 2.6–4.3 2.9–3.4 (muscle strength, skin, pulmonary function, gastrointestinal disease) and the effect of the disease on the child’s overall function. The assessment of muscle strength in children using manual muscle testing has been previously reviewed as part of a review of clinical assessment and investigation of JDM [9]. There is no recognized tool for comprehensively assessing skin disease, although it is a recognized component in the disease activity scores. It is recognized that ulcerative skin disease is a sign associated with a poor prognosis [10]. Some centers use this as one of the criteria for more aggressive treatment regimens such as the use of cyclophosphamide [11]. There has been debate about nailfold capillaroscopy in childhood rheumatic disease and the meaning of the findings in relation to disease activity [12], but more work needs to be done before its routine use in clinical assessments can be recommended. Assessment of pulmonary involvement The strength of muscles affecting speech, swallowing, and respiration also needs to be assessed. Abnormalities cannot always be elicited by questioning: swallowing problems may result in severe aspiration on videofluoroscopy and yet be asymptomatic (personal experience) [9]. Aspiration can lead to secondary lung changes that can be seen early on a high-resolution CT scan. Later, changes become apparent on chest x-ray. Chest CT scans will also pick up signs of interstitial lung disease, and lung function tests with carbon monoxide diffusion measurements are helpful [13]. MRI scan Muscle biopsies and electromyographies are considered too invasive or painful by many pediatricians, and MRI scans are used instead (Brown, Personal communication, June 2004). MRI scans do pick up muscle inflammation and demonstrate the patchy nature of the inflammation seen within muscles. It has a predictive power similar to that of muscle biopsies [14] and can distinguish between active disease and inactive disease [15]. Muscle biopsy is still considered the gold standard, but it is not always positive, even in cases in which the diagnosis is not in doubt. Recent studies have suggested that the use of immunologic stains for increased expression of the major histocompatibility complex class I protein may reduce the false-negative rate in adults [16] and children [17]. Childhood Health Assessment Questionnaire The child’s function can be assessed using the Childhood Health Assessment Questionnaire (CHAQ) [18]. This has been validated in both juvenile idiopathic arthritis [19] and JDM [18]. It is one of the most widely used health questionnaires in pediatric rheumatology and has been translated into many languages. Assessment in juvenile idiopathic inflammatory myopathies Pilkington 675 Validation of the Childhood Myositis Assessment Scale The Childhood Myositis Assessment Scale (CMAS) was developed to assess muscle strength and endurance in children with myositis [20]. It is a 14-item observational tool that is easy to use, gives a numerical score out of a total of 52, and takes approximately 10 to 15 minutes to complete. It does not require sophisticated equipment. It measures a child’s muscle strength, physical function, and endurance. Its limitations are that it is difficult to perform in young children younger than 4 years old. It can also be limited by contractures, which make some of the maneuvers difficult even when the muscle strength is normal. It has been validated in JDM [21] and has become widely used by the pediatric rheumatology community. However, more needs to be known about its properties if it is to be used as one of the standardized assessment tools by specialists and nonspecialists. Recent work has been undertaken to validate the CMAS by the Juvenile Dermatomyositis Disease Activity Collaborative Study Group, which includes pediatric rheumatologists from Canada and the United States [22••]. In this multicenter study, 108 juvenile idiopathic inflammatory myopathy patients (98 JDM, six juvenile polymyositis, and four myositis with connective tissue diseases) were assessed twice at an interval of 7 to 9 months. Within this group, 26 patients were evaluated on the same day by two examiners. The results showed that the CMAS had very good interrater reliability (intraclass correlation of 0.89) and good construct validity. Childhood Myositis Assessment Scale correlations The CMAS correlated well with the CHAQ and manual muscle testing, as was predicted: all three measure muscle strength or a function of muscle strength. The CMAS correlated moderately with assessments of overall disease activity (physician Visual Analogue Score, parental Visual Analogue Score), again as was predicted, because muscle strength does not always correlate directly with muscle inflammation or extraskeletal manifestations of disease. In fact, the data suggested that the CMAS and manual muscle testing were nonredundant (i.e., did not measure exactly the same properties), whereas the CMAS and CHAQ measure very similar properties (i.e., function), and thus one of the two may be redundant. In practice, most clinicians would expect to use all three as the CHAQ includes the parent/patient perception of well-being, and this does not always correlate directly with the clinician’s view. Parents may not realize the severity of their child’s condition at the time of diagnosis or may have difficulties in separating psychological problems from disease activity during convalescence. Conversely, clinicians may underestimate the effect of the disease on an individual child and his or her family. The CHAQ is also known to have limitations with a “floor effect”: children with no disease and children with mild disease either score 0 or close to 0. For these reasons, all three need to be included in an assessment of a patient with JDM. Whenever a unit or center takes up a new tool, it takes time for the assessors to become familiar with it and to be able to interpret what the final score actually means clinically and what constitutes a clinically meaningful change. An important aspect of this work was to look at the “minimum clinically important difference,” and to evaluate the correspondence between scores and physical dysfunction. This was only preliminary work, but it suggested that a change in CMAS score of 1.5 correlated with a change in CHAQ score of 0.13. The corresponding CHAQ values and disability levels taken from a juvenile idiopathic arthritis study [23] are shown in Table 2. Disease activity tools Disease Activity Score A disease activity tool needs to evaluate all the organs that can be affected by the disease. In both adults and children, dermatomyositis primarily affects the muscles and the skin. However, it can be a multisystem disease with widespread vasculopathy including the gastrointestinal tract, lungs, and central nervous system. This diversity is seen more often in children than in adult patients. Until recently, there were no widely available disease activity assessment tools. Some centers developed their own, and the one used by the Chicago group (Division of Immunology/Rheumatology at Northwestern’s Children’s Memorial Hospital) was published in early 2003 [24•]. This has been called the Disease Activity Score and gives a score from 0 to 20. The items evaluating muscle and skin disease are given equal weighting. The score does not include any serologic markers or any evaluation of gastrointestinal, pulmonary, or neurologic problems. It can be done in any clinical setting because it does not require any investigative equipment. Three pediatric rheumatologists assessed 44 patients with a total of 58 visits. At one visit, two of the three clinicians separately assessed 29 patients, and on a further visit, one of the original two clinicians and a third Table 2. Relationship between CHAQ values and physical disability in JIA and corresponding CMAS values in JDM CHAQ values (95% CI) Physical disability in JIA Equivalent CMAS scores (95% CI) 0.00 (−0.07, 0.07) 0.24 (0.14, 0.35) 0.71 (0.52, 0.91) 1.53 (0.96, 2.10) None Mild Mild-moderate Moderate 48 (47.2–48.8) 45 (43.7–46.3) 39 (36.4–41.6) 30 (22.8–37.2) CHAQ, Childhood Health Assessment Questionnaire; CMAS, Childhood Myositis Assessment Scale; JIA, juvenile idiopathic arthritis. 676 Myositis and myopathies clinician again assessed 29 patients (some patients being present at both visits). The estimated disease activity measures were affected by the sensitivity of the individual raters to the presence or absence of skin signs. This suggests the need for clearer descriptions of the skin indicators to improve interrater reliability. International Myositis Assessment and Clinical Studies Group The International Myositis Assessment and Clinical Studies Group brought together international experts from the adult and pediatric arenas to develop disease activity and damage tools for myositis using the experience gained from the development of similar tools in patients with lupus erythematosus. Two activity and two damage tools [25•,26] were produced: the Myositis Intention to Treat Activity Index and the Myositis Disease Activity Assessment (by Visual Analogue Scale) for disease activity, with the Myositis Damage Index and the Myositis Disease Damage Assessment (by Visual Analogue Scale) for damage. The disease activity tools assess the presence and extent of activity in seven systems: constitutional, articular, cardiac, pulmonary, gastrointestinal, cutaneous, and skeletal muscle. The damage tools identify the presence and severity of damage within the systems. The reliability of the tools and interrater reliability were assessed as fair to good for initial real patient exercises involving experts in myositis and patients with myositis. These tools are now undergoing further multicenter validation studies. Paediatric Rheumatology International Trials Organisation A different approach was taken to produce core sets of measures for disease activity and damage [27••] for JDM by the Paediatric Rheumatology International Trials Organisation in collaboration with the Pediatric Rheumatology Collaborative Study Group (an American and Ca- nadian group). Two questionnaire surveys were sent to 267 clinicians from 46 countries asking them to select and rank response variables that they used for assessing clinical response. Forty experienced pediatric rheumatologists from 34 countries then attended a consensus conference. They selected the domains and variables to be included in the preliminary disease activity and damage core sets for JDM and juvenile systemic lupus erythematosus [28]. • The domains for disease activity included the physician’s global assessment (Visual Analogue Scale or Likert Scale), muscle strength assessment (CMAS or manual muscle testing), functional ability assessment (CHAQ), muscle enzymes, global assessment by parents/patients, and a global JDM disease activity tool (Disease Activity Score or Myositis Intention to Treat Index/Myositis Disease Activity Assessment Visual Analogue Scale). • The domains for disease damage included the physician’s global assessment (Visual Analogue Scale or Likert Scale), muscle strength assessment development, and a global JDM disease damage tool (Myositis Intention to Treat Index/Myositis Disease Damage Assessment Visual Analogue Scale). The activity core set is very similar to that proposed by the International Myositis and Clinical Studies Group (Table 3). However, both the activity and damage core sets include both muscle strength and functional assessments: they cannot by themselves distinguish between the two. Previous damage (such as contractures) can interfere with activity assessments [28], and clinicians can have difficulty separating activity from damage. This difficulty was thought to make a parent’s global assessment of damage too unreliable. A further complicating factor is that many features considered by adult physicians to be damage (e.g., calcinosis) are reversible in children. There- Table 3. Comparison of proposed sets of measures for disease activity in juvenile dermatomyositis from two international collaborative efforts Target population Domain Physician’s assessment Muscle strength Laboratory measurement Functional ability Patient/parent assessment Global disease activity tool Extramuscular disease PRINTO [27••] IMACS [25,26] JDM All pediatric IIM Physician’s global assessment of disease activity by VAS or Likert Scale CMAS, MMT CK, LDH, aldolase, ALT, AST CHAQ Patient/parent global assessment of overall well-being using VAS or Likert Scale DAS or MDAA — Physician’s global assessment of disease activity by VAS or Likert Scale MMT At least 2 of CK, LDH, aldolase, ALT, AST CHAQ, and CMAS — — MDAA PRINTO, Paediatric Rheumatology International Trial Organisation; IMACS, International Myositis Assessment and Clinical Studies Group; JDM, juvenile dermatomyositis; IIM, idiopathic inflammatory myopathies; VAS, Visual Analogue Scale; CMAS, Childhood Myositis Assessment Scale; MMT, manual muscle testing; CK, creatine kinase; LDH, lactate dehydrogenase; ALT, alanine aminotransferase; AST, aspartate aminotransferase; CHAQ, Childhood Health Assessment Questionnaire; DAS, Disease Activity Score; MDAA, Myositis Disease Activity Assessment (which combines the Myositis Disease Activity Assessment by VAS and the Myositis Intention to Treat Activity Index tools). Published with permission [29•]. Assessment in juvenile idiopathic inflammatory myopathies Pilkington 677 fore, a slightly different definition of damage is needed in pediatric cases. The same groups through a large-scale international collection from the follow-up of patients with JDM are now prospectively validating these core sets. Conclusion The willingness of the international community to collaborate is allowing clinical research on JDM to progress at an exciting rate. Validating assessment tools for standardized rating of the different aspects of disease activity will allow multicenter studies to flourish. Building these into tools assessing disease activity and disease damage will allow international drug trials to be undertaken. Once these tools have been validated, it will be important to move forward and conduct these studies because they will determine which treatments are the most effective and so will directly benefit patient care. 11 Riley P, Maillard SM, Wedderburn LR, et al.: Intravenous cyclophosphamide pulse therapy in juvenile dermatomyositis. A review of efficacy and safety. Rheumatology Oxford) 2004, 43:491–496. 12 Dolezalova P, Young SP, Bacon PA, et al.: Nailfold capillary microscopy in healthy children and in childhood rheumatic diseases: a prospective single blind observational study. Ann Rheum Dis 2003, 62:444–449. 13 Kobayashi I, Yamada M, Takahashi Y, et al.: Interstitial lung disease associated with juvenile dermatomyositis: clinical features and efficacy of cyclosporin A. Rheumatology Oxford) 2003, 42:371–374. 14 Kimball AB, Summers RM, Turner M, et al.: Magnetic resonance imaging detection of occult skin and subcutaneous abnormalities in juvenile dermatomyositis: implications for diagnosis and therapy. Arthritis Rheum 2000, 43:1866–1873. 15 Maillard SM, Jones R, Owens C, et al.: Quantitative assessment of MRI T2 relaxation time of thigh muscles in juvenile dermatomyositis. Rheumatology 2004, 43:603–608. 16 Civatte M, Schleinitz N, Krammer P, et al.: Class I MHC detection as a diagnostic tool in non-informative muscle biopsies of patients suffering from dermatomyositis. Neuropathol Appl Neurobiol 2003, 29:546–552. 17 Li CK, Varsani H, Holton JL, et al.: MHC class I overexpression on muscles in early juvenile dermatomyositis. J Rheumatol 2004, 31:605–609. 18 Feldman BM, Ayling-Campos A, Luy L, et al.: Measuring disability in juvenile dermatomyositis: validity of the childhood health assessment questionnaire. J Rheumatol 1995, 22:326–331. 19 Singh G, Athreya BH, Fries JF, et al.: Measurement of health status in children with juvenile rheumatoid arthritis. Arthritis Rheum 1994, 37:1761–1769. 20 Lovell DJ, Lindsley CB, Rennebohm RM, et al.: Development of validated disease activity and damage indices for the juvenile idiopathic inflammatory myopathies. II. The Childhood Myositis Assessment Scale (CMAS): a quantitative tool for the evaluation of muscle function. The Juvenile Dermatomyositis Disease Activity Collaborative Study Group. Arthritis Rheum 1999, 42:2213–2219. 21 Lovell DJ, Giannini EH, Rider L, et al.: Validation and rater reliability of the childhood myositis assessment scale for the juvenile myositis collaborative study group [abstract]. Arthritis Rheum 1996, 38(suppl):S183. 22 •• Huber AM, Feldman BM, Rennebohm RM, et al.: Validation and clinical significance of the Childhood Myositis Assessment Scale for assessment of muscle function in the juvenile idiopathic inflammatory myopathies. Arthritis Rheum 2004, 50:1595–1603. A good paper on clinical functions of CMAS. 23 Dempster H, Porepa M, Young N, et al.: The clinical meaning of functional outcome scores in children with juvenile arthritis. Arthritis Rheum 2001, 44:1768–1774. 24 • Bode RK, Klein-Gitelman MS, Miller ML, et al.: Disease activity score for children with juvenile dermatomyositis: reliability and validity evidence. Arthritis Rheum 2003, 49:7–15. Acknowledgment The author thanks Dr. L. Wedderburn and S. Arscott for their careful reading of the manuscript, V. Brown and A. Juggins for their help in preparing this manuscript, and international colleagues for their invaluable cooperation. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest 1 • Mendez EP, Lipton R, Ramsey-Goldman R, et al.: US incidence of juvenile dermatomyositis, 1995–1998: results from the National Institute of Arthritis and Musculoskeletal and Skin Diseases Registry. Arthritis Rheum 2003, 49:300–305. 2 Cassidy JT, Petty RE: Juvenile dermatomyositis. In Textbook of Pediatric Rheumatology, 4th ed. Edited by Cassidy JT, Petty RE. London: WB Saunders, 2001. 3 Bitnum S, Daeschner CW Jr, Travis LB, et al.: Dermatomyositis. J Pediatr 1964, 64:101–131. 4 Spencer CH, Hanson V, Singsen BH, et al.: Course of treated juvenile dermatomyositis. J Pediatr 1984, 105:399–408. 25 • 5 Fisler RE, Liang MG, Fuhlbrigge RC, et al.: Aggressive management of juvenile dermatomyositis results in improved outcome and decreased incidence of calcinosis. J Am Acad Dermatol 2002, 47:505–511. Isenberg DA, Allen E, Farewell V, et al.: International consensus outcome measures for patients with idiopathic inflammatory myopathies. Development and initial validation of myositis activity and damage indices in patients with adult onset disease. Rheumatology Oxford) 2004, 43:49–54. 26 6 Oddis CV, Conte CG, Steen VD, et al.: Incidence of polymyositis-dermatomyositis: a 20-year study of hospital diagnosed cases in Allegheny County, PA 1963–1982. J Rheumatol 1990, 17:1329–1334. Pilkington C, Murray KJ, Isenberg D: Development of disease activity and damage indices for myositis—initial testing of 4 tools in juvenile dermatomyositis [abstract]. Arthritis Rheum 2001, 44:S294. 7 Symmons DP, Sills JA, Davis SM: The incidence of juvenile dermatomyositis: results from a nation-wide study. Br J Rheumatol 1995, 34:732–736. 27 •• 8 Bohan A, Peter JB: Polymyositis and dermatomyositis (first of two parts). N Engl J Med 1975, 292:344–347. Ruperto N, Ravelli A, Murray KJ, et al. for the Paediatric Rheumatology International Trials Organisation (PRINTO) Pediatric Rheumatology Collaborative Study Group: Preliminary core sets of measures for disease activity and damage assessment in juvenile systemic lupus erythematosus and juvenile dermatomyositis. Rheumatology (Oxford) 2003, 42:1452–1659. 28 9 Rider LG: Assessment of disease activity and its sequelae in children and adults with myositis. Curr Opin Rheumatol 1996, 8:495–506. Rider LG, Miller FW: Classification and treatment of the juvenile idiopathic inflammatory myopathies. Rheum Dis Clin North Am 1997, 23:619–655. 10 Santmyire-Rosenberger B, Dugan EM: Skin involvement in dermatomyositis. Curr Opin Rheumatol 2003, 15:714–722. 29 • Wedderburn LR, Li CK: Paediatric idiopathic inflammatory muscle disease. Best Pract Res Clin Rheumatol 2004, 18:345–358. A good review of JDM. Use of imaging to assess patients with muscle disease David L. Scotta,b and Gabrielle H. Kingsleya,c Purpose of review A variety of imaging modalities can be used in muscle diseases. These range from plain x-rays to conventional magnetic resonance imaging (MRI) and phosphate magnetic resonance spectroscopy (MRS). This review places these imaging methods into their relevant clinical contexts on the basis of the best available research evidence. Recent findings Plain x-rays have limited roles in imaging patients with muscle disease. An exception is identifying calcinosis in patients with myositis; there is some evidence that effective early treatment may reduce its frequency and severity. Scintigraphy has been used in several centers but it appears to have limited value. Ultrasound, though successfully used in a number of units, is relatively little used, though the evidence suggests it would be sensible if this method were adopted more widely. MRI is currently the key imaging modality. It is useful in diagnosing pyomyositis, diabetic muscle infarction, and inflammatory myositis. Its main proven value is identifying the best sites for biopsy in early myositis, though it can help differentiate between different forms of muscle disease when there is diagnostic uncertainty. The area of most intense ongoing original research is MRS, which can show the bioenergetics of normal and abnormal muscles. Changes in the ratios of inorganic phosphate and phosphocreatine, particularly during exercise provide insights into the metabolic consequences of muscle diseases and may, in the future, suggest alternative therapeutic approaches. Summary Magnetic resonance imaging is a useful adjunct when diagnosing muscle diseases. It is particularly useful to identify suitable sites for muscle biopsy. Ultrasound may be equally helpful, though there is less supporting evidence from existing research. MRS is the area in which most current novel research is focused. Keywords dermatomyositis, polymyositis, ultrasound, magnetic resonance imaging, magnetic resonance spectroscopy Curr Opin Rheumatol 16:678–683. © 2004 Lippincott Williams & Wilkins. a Department of Rheumatology, GKT School of Medicine, Weston Education Centre, Kings College, London, UK; bDepartment of Rheumatology, Kings College Hospital, Denmark Hill, London, UK; and cDepartment of Rheumatology, University Hospital Lewisham, Lewisham High Street, London, UK Correspondence to David Scott, Department of Rheumatology, GKT School of Medicine, Weston Education Centre, Kings College, 10 Cutcombe Road, London SE5 9RS, UK Tel: 44 (0)207 848 5215; fax: 44 (0) 207 848 5202; e-mail [email protected] 678 Current Opinion in Rheumatology 2004, 16:678–683 © 2004 Lippincott Williams & Wilkins 1040–8711 Introduction Imaging methods currently only have subsidiary roles in the diagnosis and assessment of muscle diseases. Such limited roles contrast with their central importance in other forms of musculoskeletal disease, in particular inflammatory synovitis. This situation is likely to change in the future as technical developments in imaging methods increase their scope and value. A range of imaging methods has been used in muscle disease. Some methods, including plain x-rays, computerized tomography (CT), and scintiscans, have circumscribed roles, which are unlikely to develop significantly in the foreseeable future. Other methods, in particular ultrasound scans, magnetic resonance imaging (MRI), and the associated technique of magnetic resonance spectroscopy (MRS) are of relatively intense research interest and they are likely to develop substantially in future years. This review highlights the main uses of imaging in muscle diseases and places the most recent research into an historical context. Range of muscle diseases Myositis ossificans This disorder is characterized by muscle calcification, and usually follows trauma [1]. It mainly involves thighs and upper arms, with pain, loss of movement, and swelling. One rare variant is myositis ossificans progressiva [2], a hereditary disorder of unknown cause. Pyomyositis Abscess formation deep within large muscles [3] spreads from soft tissue infections or through the blood. It is especially a problem in immunocompromised patients. It mainly involves quadriceps and upper arms causing pain, edema, and fever. In the initial nonsuppurative phase, pyomyositis responds to antibiotics alone. When pus is present, drainage is mandatory. Muscle infarction Occasional patients with diabetes mellitus develop muscle infarction [4], especially if poorly controlled. It is characterized by sudden, severe muscle pain and tender- Imaging and muscle disease Scott and Kingsley 679 ness, sometimes with a palpable mass; the thighs are mainly involved. Treatment is symptomatic and conservative because most cases resolve spontaneously. Idiopathic inflammatory myositis Polymyositis and dermatomyositis are characterized by inflammatory and degenerative changes in the muscles and skin in dermatomyositis [5]. They result in symmetrical weakness and muscle atrophy, principally of the limb girdles. Their onset varies from acute to insidious. The first symptom is often proximal muscle weakness or rash. Muscle tenderness and pain are less marked. Rash, polyarthralgias, Raynaud’s phenomenon, dysphagia, pulmonary disease, and constitutional complaints such as fatigue often occur. Muscle weakness is only obvious when there is substantial destruction of muscle fibers. Diagnosis depends on proximal muscle weakness, the characteristic rash, elevated serum muscle enzymes, muscle biopsy changes, and electromyographic abnormalities. Inclusion body myositis is a chronic disorder of unknown cause characterized by progressive muscle weakness and wasting that usually affects older people [6,7]. Unlike dermatomyositis and polymyositis it also affects distal muscles, with weakness of wrist and finger muscles. The pathology includes inflammatory infiltrates, intracellular degenerative changes, and abnormal mitochondria, which include the presence of ragged red fibers, paracrystalline inclusions, and heterogeneous deletions of mitochondrial DNA [8,9]. The inflammation usually does not respond to conventional immunotherapy. Plain x-rays Calcinosis Subcutaneous calcinosis is uncommon; a recent survey of 35 patients with juvenile dermatomyositis identified five (14%) patients with radiologic calcinosis [10]. A retrospective review of juvenile dermatomyositis [11] showed that calcinosis was associated with delays in starting treatment, prolonged elevations of muscle enzymes, and long disease durations. Aggressive treatment that achieved rapid and complete control of muscle inflammation reduced the risk of developing calcinosis. Calcification in myositis is not just subcutaneous; one recent report described periarticular calcification in early polymyositis [12]. Myositis ossificans As imaging is an inherent part of diagnosis, it is often mentioned in case reports. Examples include the development of myositis ossificans presenting as groin pain in soccer players [13] and the apparently successful use of alendronate to treat a young man who developed myositis ossificans after strenuous physical activity [14]. Scintigraphy Several units have evaluated scintigraphy in myositis; all initially reported encouraging results. Several techniques were described including 99mTc-MDP (methylendiphosphate) scintigraphyl [15], 111-indium altumomab pentetate-labeled antimyosin [16], and 99mtechnetiumpyrophosphate muscle. However, this approach has not developed greatly. Although a further recent report describes an alternative scintigraphic approach—using Ga67 scintigraphy to evaluate polymyositis—no technique has yet been widely adopted. Ultrasound Kane et al. [17•] reviewed ultrasound in muscle diseases and other musculoskeletal conditions. They concluded it is useful in diagnosing inflammatory muscle disease, though MRI is more sensitive in detecting edema. Ultrasound is also able to image myositis ossificans, pyomyositis, and infarction in diabetes mellitus [18] together with calcium deposits in dermatomyositis. One report compared ultrasound with MRI in 12 children with pyomyositis [19•]. Both modalities showed characteristic changes of pyomyositis. Ultrasound was preferable for imaging the extremities, but in the pelvis, MRI was better. It was also best for differentiating pyomyositis from osteomyelitis. Magnetic resonance imaging and spectroscopy Proton-based MRI methods are standard and a few centers also use phosphate imaging. MRI shows inflammation, edema, fibrosis, fatty infiltration, and calcification. Degeneration of muscle fibers themselves cannot be shown directly on MRI. Muscle edema and inflammation give relatively normal appearances in T1 and protondensity weighted images and high-signal intensities on T2-weighted fat- or non-fat-saturated sequences. It may be difficult to differentiate edema from inflammation on MRI. Magnetic resonance spectroscopy (MRS) records similar data to MRI but presents it in a different manner, indicating the amount of specific chemical entities by their resonance in a defined volume of tissue. Pyomyositis One recent review [20] concluded ultrasound, CT, and MRI are all valuable in diagnosis and directing therapy, including drainage. CT is better for monitoring progress; in advanced disease, it shows the extent of destructive changes. Case reports highlight the value of CT; for example, a young boy with complement deficiency who developed pyomyositis of the psoas [21] and a teenager in the early stage of pyomyositis in whom CT showed inflammation in the sternocleidomastoid muscle [22]. Tuberculous myositis is a rare and challenging problem, and a recent review of 35 cases [23] showed that CT or MRI were identified features suggestive of tuberculous myositis in 15 patients (43%). 680 Myositis and myopathies Muscle infarction Kapur et al. [24•] reported one case and reviewed over 100 other reports, describing clinical experience in 116 cases and pathology in 78 cases of diabetic muscle infarction (Table 1). MRI findings are not pathognomonic, but there are several characteristic features. These include extensive muscle edema, muscle enlargement, subcutaneous edema, and interfascial edema. Muscle infarction is a risk in patients with diabetes who have other disorders. Lentine and Guest [25] describe it as a complication in dialysis patients with diabetic nephropathy. In these cases isolated skeletal muscle infarction was shown on MRI. Myositis Magnetic resonance imaging defines the extent of anatomic changes. In the acute phase edema is seen using T2-weighted and/or short tau inversion recovery (STIR) imaging, and there are normal images using T1-weighted imaging [26,27]. In chronic disease, when muscular atrophy and fatty infiltration within atrophied muscles are the dominant features, a combination of T1-weighted and fat-suppressed images are most relevant. MRI findings in myositis were first described in 13 patients by Kaufman et al. [28] over 15 years ago. Subsequent studies in adults [29–31] and children [32–34] confirmed the specificity of MRI changes. There is one recent report recommending whole body MRI in myositis [35]. Occasional patients with amyopathic dermatomyositis have clinical features of dermatomyositis but no clinically detectable myopathy. MRI shows some of these cases have muscle involvement. In one study of 40 Chinese patients presenting with dermatomyositis, 10 had amyopathic dermatomyositis [36]; MRI showed abnormal signal intensity in muscles on both T2- and fat suppression sequences in three cases. Schweitzer et al. [37] examined the cost effectiveness of MRI as a biopsy guide in 25 patients with suspected polymyositis; 14 had preoperative MR imaging. In the patients whose biopsy site was selected using MRI find- ings there was only one false-negative biopsy compared with five false-negative biopsies in patients without imaging. Pre-biopsy MRI was associated with reduced medical costs—$14,000 compared with $20,000—and appeared cost effective. Recent case series illustrate the benefits of MRI in myositis. Keily et al. [38] described four cases from a series of 78 patients with myositis seen at a single UK center. They all highlighted the complexities in the presentation and natural history and showed the important and growing role of MRI in routine care. Maillard et al. [39••] evaluated MRI in children, studying 10 children with active dermatomyositis, 10 with inactive disease and 20 healthy controls. MRI T2 relaxation times were increased in active dermatomyositis and MRI scores correlated with muscle strength and function, though they were unrelated to muscle enzymes. Dion et al. [40] developed diagnostic imaging criteria for polymyositis and sporadic inclusion body myositis in a series of 25 patients each with polymyositis and inclusion body myositis. MRIs were abnormal in all patients. Fatty infiltration and atrophy were frequent in inclusion body myositis. Inflammation as the sole abnormality was preferentially seen in polymyositis. Involvement of the anterior group, an asymmetric distribution, and a distal predominance were all more frequent in inclusion body myositis. These changes are summarized in Table 2. This work attracted some subsequent criticism, mainly related to the limitations of the available diagnostic criteria [41]. Mastaglia et al. [42] in a recent review concluded that MRI is helpful in confirming the diagnosis and selecting appropriate biopsy sites, though its diagnostic sensitivity is not fully evaluated. However, another review by Dalakas and Hohlfeld [43] made little mention of MRI, and relegated it to having a minor role in occasional cases to identify inflammatory sites and select the area for biopsy. It is, at present, uncertain exactly what role MRI should play in assessing myositis [44]. Table 1. MRI and pathological findings of DMI Assessment Findings MRI T2 Hyperintense signal of infarcted muscle T2 Muscle enlargement T2 Subcutaneous edema T2 Subfascial edema Muscle fiber necrosis Inflammatory cell infiltrate Microvascular abnormality Edema Hemorrhage Fibrosis/granulation Tissue-regenerating muscle fibers Histopathology Adapted from [24]. Number of cases (%) 78/78 (100%) 47/47 (100%) 38/42 (90%) 34/38 (90%) 73/75 (97%) 48/56 (86%) 54/65 (83%) 27/55 (49%) 27/56 (48%) 36/60 (60%) 33/59 (56%) Magnetic resonance spectroscopy in myositis Pfleiderer et al. [45••] reported in vivo MRS studies in 10 patients with dermatomyositis and 18 healthy controls. They evaluated short-term alterations of metabolic parameters in short cycles of submaximal exercise. Pi/PCr (inorganic phosphate/phosphocreatine) ratios increased in both patients and controls during exercise. In controls they rapidly returned to baseline values. However, the subsequent decrease during the break period was incomplete in patients with dermatomyositis (Fig. 1). Consequently, in dermatomyositis, Pi/PCr did not return to baseline levels and subsequent rises decreased with each exercise cycle. Imaging and muscle disease Scott and Kingsley 681 Table 2. MRI evaluation of 22 patients with polymyositis and 25 patients with inclusion body myositis Fatty infiltration Atrophy Inflammation Global analysis Presence of fatty infiltration Anterior group exclusively Anterior group (Grade 3 or 4) All three groups involved Fascial pattern Widespread pattern Asymmetrical Distal predominance Presence of atrophy Anterior group atrophy Undulating fascia Presence of inflammation Inflammation in posterior group Fascial pattern Widespread pattern Asymmetrical Isolated inflammation Polymyositis Inclusion body myositis Significance 68 4 20 44 28 28 4 12 64 16 32 72 32 40 16 8 20 96 40 76 44 8 88 44 64 92 60 64 72 8 20 44 10 0 <0.03 0.0021 0.001 NS NS 0.0001 0.0009 0.0002 0.02 0.0014 0.02 NS 0.03 NS 0.03 NS 0.05 Adapted from [40]. Magnetic resonance spectroscopy was used to measure urinary creatine levels in myositis by Chung et al. [46]. Urinary creatine was detected using MRS in 26 of 35 patients with myositis, 4 of 60 cases with other medical disorders, and 10 of 50 healthy controls. Urinary Figure 1. Typical Pi/PCr curves for a control case and a dermatomyositis patient in contrast to controls creatine/creatinine ratios exceeded 0.4 in 20 patients with PM/DM but were not seen in any controls. Future work Magnetic resonance spectroscopy studies of high energy phosphate metabolism are being used to understand the biology of normal muscle, and this may soon impact on imaging muscle disease. Brosseau et al. [47] studied anaerobic exercise in the dominant and nondominant forearms of sedentary subjects. MRS showed timedependent changes in Pi and PCr concentrations. Metabolic kinetics in the dominant forearms improved during repetitive high-intensity exercise compared with the nondominant forearm muscles. Their findings provide a way of investigating how training may improve muscle bioenergetics in muscle disease. Schocke et al. [48] showed PCr hydrolysis during incremental exercises in normal muscles enters a steady state at different workload levels. In normal subjects creatine ingestion reduces the initial PCr resynthesis rate after brief exercise and increases the initial PCr resynthesis rate after exhaustive exercise, probably as a consequence of mitochondrial respiration [49]. This work starts to create a rationale for using creatine supplements in muscle diseases. In myositis there was a marked increase in Pi/PCr only observed during the first exercise cycle. The subsequent decrease during the break was incomplete. Adapted from [45••]. Quantitative phosphate MRS was used to investigate muscle metabolism in vivo in 9 patients with dermatomyositis and five patients with polymyositis. Postexercise MRS showed muscle oxidative metabolism was impaired in myositis. The phosphocreatine and adenosine diphosphate recovery half-times were almost twice as long as in controls. The impairment of high-energy phosphate recovery indices in myositis patients was similar to that found in cases with a primary mitochondrial disorder [50]. 682 Myositis and myopathies Conclusion Conventional imaging currently has a relatively minor role in imaging muscle disease. In patients with a suspected muscle disease in whom there is diagnostic uncertainty either MRI or ultrasound provides useful screening tools. In early myositis the balance of evidence favors obtaining an MRI, particularly as a guide to the best site for a biopsy. In the future MRS and understanding muscle bioenergetics represent important areas in which advances are likely to occur. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest 19 Trusen A, Beissert M, Schultz G, et al.: Ultrasound and MRI features of pyomyositis in children. Eur Radiol 2003, 13:1050–1055. • A detailed retrospective analysis of ultrasound and MRI findings in 12 children with pyomyositis. 20 Wilson DJ: Soft tissue and joint infection. Eur Radiol 2004 Jan 29, Epub ahead of print. 21 Tuerlinckx D, Bodart E, de Bilderling G, Nisolle JF: Pneumococcal psoas pyomyositis associated with complement deficiency. Pediatr Infect Dis J 2004, 23:371–373. 22 Flier S, Dolgin SE, Saphir RL, et al.: A case confirming the progressive stages of pyomyositis. J Pediatr Surg 2003, 38:1551–1553. 23 Wang JY, Lee LN, Hsueh PR, et al.: Tuberculous myositis: a rare but existing clinical entity. Rheumatology (Oxford) 2003, 42:836–840. 24 Kapur S, Brunet JA, McKendry RJ: Diabetic muscle infarction: case report and review. J Rheumatol 2004, 31:190–194. • A definitive update of the literature on diabetic muscle infarction outlining experience in 116 cases. 25 Lentine KL, Guest SS: Diabetic muscle infarction in end-stage renal disease. Nephrol Dial Transplant 2004, 19:664–669. Arrington ED, Miller MD: Skeletal muscle injuries. Orthop Clin North Am 1995, 26:411–422. 26 Fraser DD, Frank JA, Dalakas M, et al.: Magnetic resonance imaging in the idiopathic inflammatory myopathies. J Rheumatol 1991, 18:1693–1700. 2 Baysal T, Elmali N, Kutlu R, Baysal O: The stone man: myositis (fibrodysplasia) ossificans progressiva. Eur Radiol 1998, 8:479–481. 27 Fraser DD, Frank JA, Dalakas MC: Inflammatory myopathies: MR imaging and spectroscopy. Radiology 1991, 179:341–344. 3 Patel SR, Olenginski TP, Perruquet JL, Harrington TM: Pyomyositis: clinical features and predisposing conditions. J Rheumatol 1997, 24:1734–1738. 28 4 Grigoriadis E, Fam AG, Starok M, Ang LC: Skeletal muscle infarction in diabetes mellitus. J Rheumatol 2000, 27:1063–1068. Kaufman LD, Gruber BL, Gerstman DP, Kaell AT: Preliminary observations on the role of magnetic resonance imaging for polymyositis and dermatomyositis. Ann Rheum Dis 1987, 46:569–572. 29 5 Rider LG, Miller FW: Idiopathic inflammatory muscle disease: clinical aspects. Baillieres Best Pract Res Clin Rheumatol 2000, 14:37–54. Lamminen AE: Magnetic resonance imaging of primary skeletal muscle diseases: patterns of distribution and severity of involvement. Br J Radiol 1990, 63:946–950. 6 Askanas V, Engel WK: New advances in the understanding of sporadic inclusion-body myositis and hereditary inclusion-body myopathies. Curr Opin Rheumatol 1995, 7:486–496. 30 Fujino H, Kobayashi T, Goto I, Onitsuka H: Magnetic resonance imaging of the muscles in patients with polymyositis and dermatomyositis. Muscle Nerve 1991, 14:716–720. 7 Griggs RC, Askanas V, DiMauro S, et al.: Inclusion body myositis and myopathies. Ann Neurol 1995, 38:705–713. 31 Chung YL, Smith EC, Williams SCR, et al.: In vivo proton magnetic resonance spectroscopy in polymyositis and dermatomyositis: A preliminary study. Eur J Med Res 1997, 2:483–487. 8 Oldorfs A, Larsson NG, Lindberg C, Holme E: Mitochondrial DNA deletions in inclusion body myositis. Brain 1993, 116:325–336. 32 Keim DR, Hernandez RJ, Sullivan DB: Serial magnetic resonance imaging in juvenile dermatomyositis. Arthritis Rheum 1991, 34:1580–1584. 9 Santorelli FM, Sciacco M, Tanji K, et al.: Multiple mitochondrial DNA deletions in sporadic inclusion body myositis: a study of 56 patients. Ann Neurol 1996, 39:789–795. 33 Hernandez RJ, Keim DR, Chenevert TL, et al.: Fat-suppressed MR imaging of myositis. Radiology 1992, 182:217–219. 10 Sallum AM, Kiss MH, Sachetti S, et al.: Juvenile dermatomyositis: clinical, laboratorial, histological, therapeutical and evolutive parameters of 35 patients. Arq Neuropsiquiatr 2002, 60:889–899. 34 Kimball AB, Summers RM, Turner M, et al.: Magnetic resonance imaging detection of occult skin and subcutaneous abnormalities in juvenile dermatomyositis: implications for diagnosis and therapy. Arthritis Rheum 2000, 43:1866–1873. 11 Fisler RE, Liang MG, Fuhlbrigge RC, et al.: Aggressive management of juvenile dermatomyositis results in improved outcome and decreased incidence of calcinosis. J Am Acad Dermatol 2002, 47:505–511. 35 Powell T, Brennan D, Lynch T, et al.: Whole-body MR imaging in the diagnosis of polymyositis. AJR Am J Roentgenol 2002, 179:967–971. 36 12 Marie I, Fournet P, Janvresse A, et al.: Periarticular calcifications and arthropathy as the first manifestation of polymyositis. Clin Exp Rheumatol 2003, 21:681–682. Lam WW, Chan H, Chan YL, et al.: MR imaging in amyopathic dermatomyositis. Acta Radiol 1999, 40:69–72. 37 Schweitzer ME, Fort J: Cost-effectiveness of MR imaging in evaluating polymyositis. Am J Roentgenol 1995, 165:1469–1471. Cetin C, Sekir U, Yildiz Y, et al.: Chronic groin pain in an amateur soccer player. Br J Sports Med 2004, 38:223–224. 38 Kiely PD, Heron CW, Bruckner FE: Presentation and management of idiopathic inflammatory muscle disease: four case reports and commentary from a series of 78 patients. Rheumatology (Oxford) 2003, 42:575–582. 1 13 14 Ben Hamida KS, Hajri R, Kedadi H, et al.: Myositis ossificans circumscripta of the knee improved by alendronate. Joint Bone Spine 2004, 71:144–146. 15 Nakayama T, Saitoh Y, Yatabe K, et al.: Diagnosis of inflammatory myopathy; usefulness of 99mTc MDP scintigraphy and muscle MRI for determination of affected sites. Clin Neurol 1999, 39:1114–1117. 16 Lofberg M, Liewendahl K, Lamminen A, et al.: Antimyosin scintigraphy compared with magnetic resonance imaging in inflammatory myopathies. Arch Neurol 1998, 55:987–993. Kane D, Grassi W, Sturrock R, Balint PV: Musculoskeletal ultrasound–a state of the art review in rheumatology. Part 2:Clinical indications for musculoskeletal ultrasound in rheumatology. Rheumatology (Oxford) 2004, 43:829–838. An excellent overview of musculoskeletal ultrasound. 17 • 18 Chason DP, Fleckenstein JL, Burns DK, Rojas G: Diabetic muscle infarction: radiologic evaluation. Skeletal Radiol 1996, 25:127–132. Maillard SM, Jones R, Owens C, et al.: Quantitative assessment of MRI T2 relaxation time of thigh muscles in juvenile dermatomyositis. Rheumatology (Oxford) 2004, 43:603–608. A detailed evaluation of MRI in juvenile dermatomyositis. Ten children with active myositis, 10 with inactive disease, and 20 healthy children were studied. The MRI T2 relaxation times were significantly increased in active myositis compared with inactive disease and healthy children. There were also good correlations between the MRI scores and the measures of muscle strength and function; however, there was no correlation between the MRI and muscle enzymes. 39 •• 40 Dion E, Cherin P, Payan C, et al.: Magnetic resonance imaging criteria for distinguishing between inclusion body myositis and polymyositis. J Rheumatol 2002, 29:1897–1906. 41 Hengstman GJ: Magnetic resonance imaging criteria to differentiate inclusion body myositis from polymyositis. J Rheumatol 2003, 30:1892. Imaging and muscle disease Scott and Kingsley 683 42 Mastaglia FL, Garlepp MJ, Phillips BA, Zilko PJ: Inflammatory myopathies: clinical, diagnostic and therapeutic aspects. Muscle Nerve 2003, 27:407– 425. 43 Dalakas MC, Hohlfeld R: Polymyositis and dermatomyositis. Lancet 2003, 362:971–982. 44 Isenberg DA, Allen E, Farewell V, et al.: International Myositis and Clinical Studies Group (IMACS). International consensus outcome measures for patients with idiopathic inflammatory myopathies. Development and initial validation of myositis activity and damage indices in patients with adult onset disease. Rheumatology (Oxford) 2004, 43:49–54. Pfleiderer B, Lange J, Loske KD, Sunderkotter C: Metabolic disturbances during short exercises in dermatomyositis revealed by real-time functional 31P magnetic resonance spectroscopy. Rheumatology (Oxford) 2004, 43:696–703. 31 P MRS was used to assess short-term alterations of metabolic dynamics during muscular exercise in 10 patients with dermatomyositis and 18 healthy subjects. In five short (1 minute) cycles of submaximal exercise Pi/PCr ratios during exercise increased in patients and controls. They rapidly returned to baseline values in the 45 •• controls, but both Pi and PCr remained above baseline values in patients and resulted in irregular Pi/PCr ratios. 46 Chung YL, Wassif WS, Bell JD, et al.: Urinary levels of creatine and other metabolites in the assessment of polymyositis and dermatomyositis. Rheumatology (Oxford) 2003, 42:298–303. 47 Brosseau OE, Mahdjoub R, Seurin MJ, et al.: Kinetics of anaerobic metabolism in human skeletal muscle: influence of repetitive high-intensity exercise on sedentary dominant and non-dominant forearm. A 31P NMR study. Biochimie 2003, 85:885–890. 48 Schocke MF, Esterhammer R, Kammerlander C, et al.: High-energy phosphate metabolism during incremental calf exercise in humans measured by 31 phosphorus magnetic resonance spectroscopy (31P MRS). Magn Reson Imaging 2004, 22:109–115. 49 Smith SA, Montain SJ, Zientara GP, Fielding RA: Use of phosphocreatine kinetics to determine the influence of creatine on muscle mitochondrial respiration: an in vivo 31P-MRS study of oral creatine ingestion. J Appl Physiol 2004, 96:2288–2292. Is it really myositis? A consideration of the differential diagnosis Niranjanan Nirmalananthana, Janice L. Holtonb and Michael G. Hannaa,c Purpose of review The idiopathic inflammatory myopathies are an important and treatable group of disorders. However, the potential toxicity associated with the immune therapeutic regimens used to treat these disorders may be significant; therefore, accurate diagnosis before such treatment is essential. The differential diagnosis is potentially large. Accurate diagnosis usually depends on a combination of careful clinical assessment in conjunction with detailed laboratory investigations. Muscle biopsy remains essential in achieving an accurate diagnosis that will then guide treatment. This review describes the diagnostic approach used. Recent findings There has been debate over the requirements for an accurate diagnosis of inflammatory myopathy (i.e., polymyositis and dermatomyositis). It is increasingly recognized that there can be clinical and muscle histopathologic overlap between the features of inflammatory myopathies and those of other muscle disorders, in particular, the genetic muscular dystrophies. Pathologic findings of inflammation and major histocompatibility complex upregulation, although typical of inflammatory myopathies, have been shown to occur in some muscular dystrophies, complicating the diagnostic process. Inclusion body myositis is much less responsive to immunotherapy and is now recognized as the most common acquired muscle disease in those older than 50 years of age. It is likely that genetic muscular dystrophies and inclusion body myositis account for some cases of apparently “treatment-resistant” myositis. Summary A thorough clinical assessment, including a detailed family history, complemented by electromyography and creatine kinase measurements, should be undertaken in any patient with presumed idiopathic inflammatory myopathy. In addition, a muscle biopsy remains essential in all cases. A precise tissue diagnosis confirming features of an active inflammatory process should be achieved before immunosuppressive treatment is commenced. An increasing array of immunocytochemical and histioenzymatic stains now allows a full analysis and will help to confirm or exclude virtually all the differential diagnostic possibilities considered in this review. Electron microscopy may also be valuable in selected cases. Close collaboration between clinicians and muscle pathologists is essential in allowing the most accurate interpretation of myopathologic findings in the clinical context. Keywords myositis, inflammatory myopathies, muscular dystrophies, diagnosis Curr Opin Rheumatol 16:684–691. © 2004 Lippincott Williams & Wilkins. 684 a Neurogenetics Group, bDivision of Neuropathology, cCentre for Neuromuscular Disease; Department of Molecular Neuroscience, Institute of Neurology; and National Hospital for Neurology and Neurosurgery, London, United Kingdom Correspondence to Michael G. Hanna, MD, Centre for Neuromuscular Disease, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK Tel: 020 7837 3611; fax: 020 7692 1208; e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:684–691 Abbreviations BMD CK DM DMD IBM IIM LGMD MHC PM Becker muscular dystrophy creatine kinase dermatomyositis Duchenne muscular dystrophy inclusion body myositis idiopathic inflammatory myopathy limb-girdle muscular dystrophy major histocompatibility complex polymyositis © 2004 Lippincott Williams & Wilkins 1040–8711 Introduction Myositis is strictly a term that is used to describe infection or inflammation of skeletal muscle. In clinical practice, the term is often used synonymously with the socalled idiopathic inflammatory myopathies (IIMs), a group of disorders characterized clinically by muscle weakness (principally proximal) and fatigue and pathologically by mononuclear inflammatory infiltrates in muscle [1]. The main clinical entities in the group are dermatomyositis (DM) and polymyositis (PM) [2]. They are, by definition, considered primary autoimmune diseases directed at as yet unidentified antigens within skeletal muscle. Inclusion body myositis (IBM) had previously been considered another member of the group of IIMs, but most investigators now consider this to be a primary degenerative disease of muscle in which there may be secondary inflammatory changes [3•]. Making a diagnosis of PM and DM is essential because of the treatability of these disorders, their association with malignancy and autoimmune rheumatic disorders, and the frequency of multisystem involvement. There are, however, a number of myopathic and neurogenic disorders that may cause diagnostic difficulty. It is essential that these disorders be differentiated from IIM, particularly in view of the potential toxicity of immunosuppressive therapy. This review summarizes the clinical features of IIM, discusses common pitfalls in diagnosis, and briefly considers some of the conditions that may cause diagnostic confusion. Is it really myositis? Nirmalananthan et al. 685 Clinical features of inflammatory myopathies The clinical features of the inflammatory myopathies have been reviewed elsewhere [4,5,6••]. PM and DM are typically of subacute onset and are characterized by the development of progressive, symmetric, and usually painless, predominantly proximal muscle weakness. The weakness generally occurs earlier and is more severe in the pelvic girdle than the shoulder girdle. IBM is a more chronic disease, and diagnosis can be delayed by a number of years after symptom onset. The diagnosis of DM is established by the presence of weakness associated with a rash on sun-exposed parts, elevation of creatine kinase (CK) activity, myopathic electromyographic findings, and a distinctive histopathologic picture. Although the onset is typically subacute, it can sometimes develop acutely over days. Presentation without a rash or with a typical rash but no apparent muscle pathology may occur rarely. The presence of the rash, however, is virtually pathognomonic of the condition. In children with the rash and muscle weakness, it may be reasonable not to consider a muscle biopsy in favor of an MRI scan, but we always perform muscle biopsies in our adult [older than 16 years] population. Inclusion body myositis has a more chronic course, and a selective and asymmetric pattern of muscle involvement not usually seen in PM or DM may be helpful diagnostically. Patients typically develop wasting of the long finger flexors and the quadriceps, resulting in frequent falls. CK elevation is relatively modest and histology is distinctive. It is also resistant to therapy with conventional immunosuppressive treatments [6••]. Polymyositis is often the most diagnostically challenging because it lacks characteristic cutaneous manifestations (compared with DM), a unique distribution of weakness (compared with IBM) or a completely specific myopathologic appearance. There has recently been much debate regarding its diagnosis and differentiation from IBM [4,7••]. The most widely used criteria for the diagnosis of PM and DM are those of Bohan and Peter [8]. However, in the 1977 study of Bohan et al. [9], proximal muscle weakness was the presenting symptom in only 69% of patients, CK levels were normal in 5%, and more than 10% had a normal electromyogram, with many more lacking the typical triad of features described below. Furthermore, 12.5% of muscle biopsy samples revealed no abnormalities and were atypical in many other patients. Other more specific criteria have recently been proposed [4,10]. Depending on the individual case, the history should be directed to exclude specific alternative diagnoses, with particular attention to the family history and medication history. The standard supportive laboratory investigations merit further consideration. Serologic tests Although aspartate and alanine aminotransferases, lactate dehydrogenase, and aldolase levels are elevated in IIM, the most widely used muscle enzyme assay is CK. This can be elevated as much as 50-fold in PM and DM but rarely much higher; serum CK that is elevated more than 100-fold should call the diagnosis into question. In IBM, the CK is more mildly elevated, as much as fivefold. CK levels may, however, rarely be normal in IIMs, even in the presence of inflammatory changes found on biopsy. The explanation for this is unclear but emphasizes the importance of undertaking muscle biopsy and not relying on CK for diagnostic purposes [11,12]. CK levels may also fluctuate from day to day (increasing significantly after major exercise), even in the absence of any intervention. Furthermore, CK elevation is nonspecific, merely indicating the presence of muscle damage, and should never be regarded as a diagnostic test. CK is elevated in muscular dystrophies and in some metabolic myopathies (particularly if there is any degree of rhabdomyolysis) and although the degree of elevation can be informative, there is considerable overlap. A search for autoantibodies may be diagnostically useful in PM and DM and provide a clue to disease subtype. Indeed, the absence of a positive antinuclear antibody and an anti-Jo antibody should also raise doubts about the diagnosis. Although autoantibodies have been found in IBM [13,14], they are unusual. They are not associated with muscular dystrophies or metabolic myopathies, although their presence should not exclude these diagnoses [15]. The presence of acetylcholine receptor antibodies points to a diagnosis of myasthenia gravis. Electromyography Electromyographic findings in IIM are not specific and are useful only insofar as they confirm an active myopathic process. In PM and DM, there is evidence of increased membrane irritability such as positive sharp waves, fibrillation potentials, and complex repetitive discharges. Myopathic motor unit action potentials that are polyphasic and of short duration and low amplitude are seen. Finally, there is early or rapid recruitment of motor unit action potentials. In IBM, there may be additional evidence of neurogenic changes with prolonged, large-amplitude motor unit action potentials. This can lead to diagnostic confusion with motor neuron disease [16]. Muscle biopsy A definitive diagnosis of IIM relies on muscle biopsy, and erroneous interpretation of a muscle biopsy specimen is probably the most common cause of a clinical 686 Myositis and myopathies misdiagnosis of IIM [17]. There are many pitfalls in both the analysis and interpretation of a muscle biopsy specimen, and these have been reviewed [17]. The key myopathologic feature of PM is considered to be endomysial lymphocytic infiltration. However, similar infiltration has been reported in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) [18], facioscapulohumeral dystrophy [19], limbgirdle muscular dystrophy (LGMD) type 2B [20], and congenital muscular dystrophy with primary merosin deficiency [21] as well as in IBM. It is therefore very important to also perform appropriate immunocytochemical staining in all cases to assess for deficiency of any of the known proteins that cause muscular dystrophies. In addition, genetic testing can be very helpful, such as the genetic test that is now widely available for facioscapulohumeral dystrophy. In IBM, there are additionally Congo red–positive amyloid deposits and rimmed vacuoles that represent an important diagnostic clue, and filamentous inclusions are usually present on electron microscopy. The key myopathologic feature in DM is perivascular B cell–predominant inflammation associated with microinfarcts and perifascicular atrophy. Muscle inflammation can, however, be patchy and is affected by the early use of steroids [22]. In cases in which typical changes are not found, particular care must be taken to exclude other possible diagnoses, and immunohistochemistry and enzyme studies should be undertaken on biopsy samples. Major histocompatibility complex (MHC) class I proteins are not usually expressed on muscle fibers. However, in IIM, MHC class I is detectable by immunohistochemistry [23,24]. MHC class I is present not only on degenerating fibers but also in apparently normal fibers and in areas without overt inflammation. For this reason, it has been suggested that immunohistochemical evidence of MHC upregulation be included in the diagnostic criteria for IIM [4]. Although MHC upregulation is very helpful, it is not specific for IIMs. MHC class I upregulation, together with inflammatory infiltration, may also be found in muscle from patients with dysferlinopathies and DMD, and, as in IIM, MHC is present on normal as well actively degenerating fibers [25•]. Interestingly, it has been observed that conditional upregulation of MHC class I in mouse skeletal muscle is sufficient to cause autoimmune myositis [26]. Examples of histopathologic findings in IIMs, IBM, and muscular dystrophy are shown in Figure 1. Figure 1. Inflammatory changes can be found in muscle biopsy specimens from patients with a number of conditions including the idiopathic inflammatory myopathies and some types of muscular dystrophy In polymyositis, the inflammatory cell infiltrate is predominantly endomysial (A) with infiltration of intact myofibers (arrow). CD8-positive T lymphocytes are the dominant cell type (B) and can be seen within nonnecrotic fibers (arrow). There is widespread expression of major histocompatibility complex (MHC) class I at the periphery of myofibers (C). Inclusion body myositis is characterized by the presence of rimmed vacuoles (D, arrow) that on ultrastructural examination are found to contain whorled membranous material (E) and randomly oriented 12- to 18-nm diameter fibrils (F). In dermatomyositis, the lymphocytic infiltrate is often perivascular in distribution (G, arrow), although it extends into the endomysium. Ultrastructural examination shows a variety of pathologic findings in the capillaries including empty loops of basal lamina indicating capillary loss (H) and the characteristic tubuloreticular inclusions in endothelial cells (I). Inflammation may be a feature of dysferlinopathy (J) in which the normal sarcolemmal distribution of dysferlin immunohistochemical staining (K) is absent (L). Hematoxylin and eosin (A, D, G, J); CD8 immunohistochemistry (B); MHC class I immunohistochemistry (C); electron microscopy (E, F, H, I); dysferlin immunohistochemistry (K, L). Original magnifications: ×40 (A, G, J); ×80 (B, C, D, K, L); ×5000 (E, H); ×1600 (F); ×12,000 (I). Is it really myositis? Nirmalananthan et al. 687 Consideration of the differential diagnosis A broad differential diagnosis is presented in Table 1. In the presence of a typical rash in association with the other clinical features of DM, the diagnosis is often straightforward. Difficulties arise in patients with suspected PM or with DM without dermatitis. It is also important to review the diagnosis in patients who were considered to have an IIM but who have not responded to immunotherapy and are often labeled as having treatment-resistant PM or DM. Alternative diagnoses should also be systematically excluded in patients with atypical investigation results, especially in patients with normal muscle biopsy specimens. A few specific disorders are briefly considered here. Muscular Dystrophies Limb-girdle muscular dystrophy The LGMDs are a heterogeneous group of disorders presenting with a face-sparing, predominantly proximal, progressive muscular weakness associated with elevated muscle enzyme levels and dystrophic features on biopsy specimens (i.e., degeneration and regeneration of muscle fibers) [27]. In recent years, there have been many gene discoveries [28•,29••]. There are at least 10 recessive forms, classed as type 2 LGMD, constituting 90% of cases [29••]. The proteins implicated are diverse and include sarcolemmal components responsible for membrane stabilization, proteases, nuclear membrane proteins, and others. The distribution of weakness is often similar to that of IIM [30], and dysphagia may also occur [31]. Specific clinical features vary between subtypes [29••]; for example, the sarcoglycanopathies (LGMDs 2C to 2F) pre- Table 1. Differential diagnosis of inflammatory myopathy Muscular dystrophies, in particular: Limb-girdle muscular dystrophy, especially type 2B (dysferlinopathy) Miyoshi myopathy (dysferlinopathy) Dystrophinopathy (Becker muscular dystrophy, isolated female manifesting carriers of dystrophinopathy) Facioscapulohumeral dystrophy Metabolic myopathies, in particular: Myophophorylase deficiency (McArdle disease) Phosphofructokinase deficiency Acid maltase deficiency Mitochondrial myopathy Endocrine myopathies Drug-induced myopathy D-penicillamine Quinidine Procainamide -hydroxy--methylglutaryl-coenzyme A reductase inhibitors (statins) Interferon alpha Interleukin-2 Motor neuron disease Spinal muscular atrophy (late-onset forms) Myasthenia gravis sent very much like the dystrophinopathies, with cardiomyopathy and calf hypertrophy. Dysferlinopathy: limb-girdle muscular dystrophy 2B and Miyoshi myopathy LGMD2B and Miyoshi myopathy are caused by mutations in the dysferlin gene. Dysferlin is an integral sarcolemmal protein believed to be involved in membrane fusion and repair [32••,33]. Dysferlinopathy is an autosomal recessive condition that is clinically heterogeneous [34], even in cases with identical genetic defects [35]. It usually presents in early adulthood with weakness in a proximal (LGMD2B), proximal-distal, or distal (Miyoshi myopathy) distribution. Miyoshi myopathy starts distally in the legs, particularly in gastrocnemius and soleus muscles. Proximal progression to the pelvic girdle and the upper limbs then occurs, although the small muscles of the hand are spared. LGMD2B also usually affects the lower limbs years before the upper limbs and, as in IBM, the quadriceps muscle is often weaker than the hip muscles. Clinical clues include the fact that periscapular muscles are usually relatively spared. Creatine kinase levels are always elevated, even in the preclinical stages, and can often be much higher than in IIM with levels in the tens of thousands. Electromyography is myopathic. Muscle histology can also be confusing. Patients with both LGMD2B and Miyoshi myopathy may have significant inflammation demonstrated on muscle biopsy samples [36,37] both endomysially and perivascularly [25•]. Muscle fibers also aberrantly express MHC class I, as in IIM [25•]. The key to diagnosis is immunohistochemistry findings that demonstrate an absence of sarcolemmal dysferlin. Genetic diagnosis is difficult due to the very large size of the gene (150 kb over 55 exons). Interestingly, the SJL/J mouse, which has been used as a model of autoimmune myositis [38], has been found to have a mutation in dysferlin, resulting in greatly reduced dysferlin expression [39]. Dystrophinopathy Duchenne muscular dystrophy is the most common of the dystrophies and results from mutations in the plasma membrane–associated protein dystrophin [40••]. DMD rarely poses diagnostic problems, but other presentations of dystrophinopathy may be more difficult to differentiate from IIM. Becker muscular dystrophy Becker muscular dystrophy usually results from dystrophin mutations that result in abnormal but at least partly functional protein, whereas in DMD, there is loss of dystrophin expression. The age at onset in BMD is later than that in DMD, usually between the ages of 5 and 15 years, although it can present as late as the fourth de- 688 Myositis and myopathies cade. The pattern of wasting is very similar to that of DMD, but the severity is usually much less. Pelvic girdle and thigh muscles are involved first, with relatively early calf pseudohypertrophy. As with LGMD2B, confusion can arise with IBM due to frequent prominent involvement of the quadriceps muscle. Shoulder girdle weakness usually develops subsequently. Cardiac disease and mental retardation are rarer than in DMD, and this makes differentiation from IIM more difficult. Dystrophinopathies may be responsive to corticosteroids [41], and muscle biopsy sample can show mononuclear infiltrates, giving rise to further diagnostic difficulty. The family history of X-linked inheritance can help clarify the diagnosis, but approximately one third of cases represent new mutations. Ninety-eight percent of mutations can be detected by a multiplex polymerase chain reaction screening 19 exons of the dystrophin gene, one of the largest in the genome [40••]. Diagnosis can also be confirmed by immunostaining muscle biopsy specimens for dystrophin. The protein is absent in DMD, but in BMD, although it is present, there is usually only partial sarcolemmal staining. The immunohistochemistry findings may, however, be normal. Western blot for dystrophin in muscle allows the determination of both the quantity and size of the molecule, reduced in 80% of patients with BMD and increased in approximately 5%. Fifteen percent of BMD patients have normal-size protein of reduced quantity. Female carriers of dystrophinopathy Because of lyonization (the random inactivation of one X chromosome during early development), most female carriers of a dystrophin mutation will switch off production of the mutant gene in 50% of chromosomes and express enough normal dystrophin from the remainder to prevent phenotypic expression. In some cases, however, nonrandom inactivation results in significantly reduced dystrophin levels and phenotypic expression [42]. Muscle weakness in female carriers occurs in approximately 19% of families with DMD and 14% of families with BMD [43]. Manifesting female carriers present from their late teens onward, with progressive proximal weakness of variable severity. The inflammation seen in DMD and BMD is, however, usually absent, and muscle biopsy samples reveal scattered muscle fibers with dystrophin levels reduced or absent on immunohistochemistry. Facioscapulohumeral dystrophy Facioscapulohumeral dystrophy is the third most common muscular dystrophy after DMD and myotonic dystrophy. Selective weakness is the main distinguishing feature. Patients commonly present with onset in the face, and subsequent periscapular and humeral weakness. Later progression to the lower limbs is seen, par- ticularly distally, the reverse of the progression in the IIMs. A significant minority of muscle biopsy specimens from patients with facioscapulohumeral dystrophy show inflammatory change [19]. Almost all patients with facioscapulohumeral dystrophy harbor deletions of a tandem repeat, termed D4Z4, on chromosome 4q, and genetic diagnosis is available. Interestingly, there is no gene known at this locus, and it appears that the deletion modulates expression of more proximal genes on chromosome 4, an effect termed positional variegation [44]. Metabolic myopathies Defects in many aspects of cellular metabolism can cause myopathy. Genetic metabolic myopathies present from anytime in childhood to adulthood and tend to be slowly progressive. It is unusual for the metabolic myopathies to show the classic electromyographic triad described for PM and DM. Furthermore, a biopsy specimen does not demonstrate inflammation, and with appropriate histioenzymatic staining, a specific metabolic defect can often be identified. Sometimes there is diagnostic confusion if there has been rhabdomyolysis. In this setting, the biopsy specimen may appear to show an inflammatory infiltrate, but careful analysis usually reveals that it is only necrotic fibers that are surrounded by inflammatory cells and macrophages. In contrast, in IIMs, nonnecrotic fibers are the subject of inflammatory attack. Several metabolic myopathies present with fixed or progressive proximal muscle weakness. The main classes are muscle glycogenoses, lipid storage disorders, and mitochondrial myopathies. Muscle glycogenoses The glycogenoses (glycogen storage diseases) are autosomal recessive enzyme deficiencies impairing glycogen metabolism. They may present with either a hepatic or muscle phenotype. The most common of the muscle glycogenoses is McArdle disease (type 5 glycogenosis) caused by a deficiency of myophosphorylase. Onset is usually in early adulthood, typically with myalgia occurring soon after starting exercise. Extreme exertion may result in myoglobinuria. Some patients present late with a relatively fixed proximal myopathy, usually with a history of fatigue and exercise intolerance. Screening is by the forearm lactate test in which the normal increase in muscle lactate levels caused by repeated exercise is abolished. The nonischemic version of the test has been shown to be as effective as the ischemic lactate test and is less painful [45]. False positives are frequent, so the result must be confirmed by biochemical analysis of muscle enzyme activity or histochemical staining for myophosphorylase on muscle biopsy. A genetic test is also available. Is it really myositis? Nirmalananthan et al. 689 Muscle phosphofructokinase deficiency (Tarui disease/type 7 glycogenosis) usually causes exerciseinduced myalgia similar to McArdle disease, but a minority of patients present with a late-onset proximal myopathy [46,47], sometimes with no history of exercise intolerance. The adult-onset form of acid maltase deficiency (type 2 glycogenosis) causes proximal muscle weakness that is greater in the pelvic than the shoulder girdle and can be mistaken clinically for PM or LGMD. CK levels are elevated in almost all cases. Electromyography shows nonspecific myopathic changes but may be normal in as many as 25% of patients, and the muscle biopsy specimen usually shows lysosomal vacuolation but again may be normal in as many as 25% of patients [48]. Clinically, a major clue is relatively early diaphragmatic involvement [49]. The rarer brancher deficiency glycogenosis (type 4 glycogenosis) may also present as progressive proximal myopathy [50], although this is usually a rapidly progressive juvenile disease with marked hepatic involvement. Lipid storage disorders Carnitine palmitoyl transferase II deficiency is the most common of the lipid storage disorders. It usually manifests as muscle pain induced by prolonged exercise. Myoglobinuria is frequent. However, some patients present with a painless proximal myopathy. Muscle biopsy specimens show abnormal lipid accumulations, and muscle tissue can be used for specific enzyme assays. Other rarer lipid storage disorders can also present with proximal myopathy, including primary carnitine deficiency [51], which is easily treatable with carnitine supplementation. Mitochondrial myopathies Mitochondrial disease is very heterogeneous and can present in many ways including ophthalmoplegia, stroke, and epilepsy [52]. Mitochondrial myopathy usually presents as a symmetric proximal myopathy associated with fatigue, much like IIM. Clinical clues prompting investigation of mitochondrial disease are few, and a detailed history and examination are relied on to find associated features such as diabetes or evidence of a family history of features consistent with those of mitochondrial disease such as deafness, diabetes, and developmental delay. Pedigrees may demonstrate maternal inheritance in the case of mitochondrial DNA disorders, but nuclear mitochondrial diseases are inherited in a Mendelian fashion. The electromyogram is often normal. Diagnosis relies on a combination of clinical findings, muscle histology, biochemical studies, and molecular genetics [53•]. Muscle biopsy is the crucial part of the investigation, both for positive identification and for differentiation from other proximal myopathies. However, classic histologic features such as the presence of ragged red fibers are not entirely specific and may be seen in IBM or acid maltase deficiency, nor does their absence exclude mitochondrial myopathy. Endocrine myopathies A number of endocrinopathies are associated with proximal myopathy [54]. The features are summarized in Table 2. Thyroid and parathyroid dysfunction is easily screened by checking T4, thyroid-stimulating hormone, calcium, and phosphate levels. Cushing syndrome is usually clinically obvious from other stigmata by the time significant myopathy is evident, as is acromegaly. A history of exogenous steroid administration should always lead one to suspect steroid myopathy. In all these cases, myopathy resolves with treatment of the underlying endocrine disorder. Hypothyroidism is the most likely to mimic myositis clinically, with significantly elevated CK and inflammation in as many as 12.5% of biopsy samples [55]. Specific considerations in the differential diagnosis of inclusion body myositis Although the most common condition that IBM is mistaken for is PM, there are a number of other differential diagnostic considerations [7••]. The early-adult onset distal myopathy with rimmed vacuoles (Nonaka myopathy) is an autosomal recessive disorder that is allelic with hereditary IBM, both of which are owing to mutations in the GNE gene [56]. Initial weakness occurs in the distal leg anterior compartment, and serum CK is moderately elevated, usually no Table 2. Features of endocrinopathies associated with proximal myopathy Endocrine disorder Distribution CK Notes Hypothyroidism Hyperthyroidism Cushing syndrome/steroid myopathy Hypoparathyroidism Proximal Proximal + distal ± bulbar Proximal Proximal ↑/↑↑ N/↓ N N/mild ↑ Hyperparathyroidism Osteomalacia Acromegaly Proximal N N N/↑ Myoedema; type II atrophy on Bx, occasionally inflammatory Weakness > wasting; frequent myalgia; Bx sample normal Fibrillation potentials absent on EMG Very rarely causes myopathy; usually tetany; EMG/Bx sample normal Hyperreflexia Type II atrophy on Bx Late in disease course, when clinically obvious Proximal Bx, biopsy; EMG, electromyogram; N, normal. 690 Myositis and myopathies more than five times normal. The quadriceps muscle, often prominently affected in sporadic IBM, is typically spared. However, there is much overlap. Furthermore, in addition to the typical vacuolation of fibers found on muscle biopsy, endomysial inflammation can also be seen in distal myopathy with rimmed vacuoles [57•]. Vacuolar myopathy similar to sporadic IBM but without inflammation is also seen in distal myopathies, including Welander distal myopathy and tibial muscular dystrophy. Proximal involvement is, however, rare in Welander distal myopathy and occurs very late in tibial muscular dystrophy, and onset of both disorders typically begins in the long finger extensors (compare with finger flexor involvement in IBM). Miyoshi myopathy was discussed previously. A recent pathologic study of three cases of X-linked Emery-Dreifuss muscular dystrophy revealed an inflammatory process very similar to that of IBM [58]. However, in general X-linked Emery-Dreifuss muscular dystrophy poses little diagnostic challenge because early contractures, mainly of the elbows and ankles, are a prominent feature, and patients develop limitation of spinal flexion. Conclusion An accurate diagnosis of IIM is important because of the treatability of these conditions. Furthermore, misdiagnosis may lead to unnecessary exposure of patients to toxic immunotherapies. Many neuromuscular disorders, in particular the genetic muscular dystrophies and metabolic myopathies, may potentially mimic the myositides. Usually a detailed clinical and myopathologic evaluation will allow the correct diagnosis to be made. Lack of response to immunotherapy should always lead to a review of the diagnosis before considering further and often increasingly toxic immunotherapies. Close collaboration between clinicians and muscle pathologists is essential to achieve the optimal management in patients with IIMs. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest 1 Engel AG, Arahata K: Mononuclear cells in myopathies: quantitation of functionally distinct subsets, recognition of antigen-specific cell-mediated cytotoxicity in some diseases, and implications for the pathogenesis of the different inflammatory myopathies. Hum Pathol 1986, 17:704–721. 2 Dalakas MC: Polymyositis, dermatomyositis and inclusion-body myositis. N Engl J Med 1991, 325:1487–1498. Askanas V, Engel WK: Proposed pathogenetic cascade of inclusion-body myositis: importance of amyloid-beta, misfolded proteins, predisposing genes, and aging. Curr Opin Rheumatol 2003, 15:737–744. This is a detailed review of what is currently known of the pathogenesis of IBM. 3 • 4 Dalakas MC, Hohlfeld R: Polymyositis and dermatomyositis. Lancet 2003, 362:971–982. 5 Hilton-Jones D: Diagnosis and treatment of inflammatory muscle diseases. J Neurol Neurosurg Psychiatry 2003, 74(suppl 2):ii25–ii31. 6 Mastaglia FL, Garlepp MJ, Phillips BA, et al.: Inflammatory myopathies: clinical, diagnostic and therapeutic aspects. Muscle Nerve 2003, 27:407–425. •• A highly comprehensive recent review of the idiopathic inflammatory myopathies. 7 van der Meulen MF, Bronner IM, Hoogendijk JE, et al.: Polymyositis: an overdiagnosed entity. Neurology 2003, 61:316–321. •• A detailed clinicopathologic study of the subclassification of inflammatory myopathy by Bohan and Peter and more recent histopathologic criteria are reported. 8 Bohan A, Peter JB: Polymyositis and dermatomyositis (first of two parts). N Engl J Med 1975, 292:344–347. 9 Bohan A, Peter JB, Bowman RL, et al.: Computer-assisted analysis of 153 patients with polymyositis and dermatomyositis. Medicine (Baltimore) 1977, 56:255–286. 10 Mastaglia FL, Phillips BA: Idiopathic inflammatory myopathies: epidemiology, classification, and diagnostic criteria. Rheum Dis Clin North Am 2002, 28:723–741. 11 Gran JT, Myklebust G, Johansen S: Adult idiopathic polymyositis without elevation of creatine kinase. Case report and review of the literature. Scand J Rheumatol 1993, 22:94–96. 12 Rider LG, Miller FW: Laboratory evaluation of the inflammatory myopathies. Clin Diagn Lab Immunol 1995, 2:1–9. 13 Hengstman GJ, Ter Laak HJ, van Engelen BG, et al.: Anti-Jo-1 positive inclusion body myositis with a marked and sustained clinical improvement after oral prednisone. J Neurol Neurosurg Psychiatry 2001, 70:706. 14 Selva-O’Callaghan A, Mijares-Boeckh-Behrens T, Labrador-Horrillos M, et al.: Anti-PM-Scl antibodies in a patient with inclusion body myositis. Rheumatology (Oxford) 2003, 42:1016–1018. 15 Funauchi M, Nozaki Y, Yoo BS, et al.: A case of limb-girdle muscular dystrophy with serum anti-nuclear antibody which led to a mistaken diagnosis of polymyositis. Clin Exp Rheumatol 2002, 20:707–708. 16 Dabby R, Lange DJ, Trojaborg W, et al.: Inclusion body myositis mimicking motor neuron disease. Arch Neurol 2001, 58:1253–1256. 17 Dalakas MC: Muscle biopsy findings in inflammatory myopathies. Rheum Dis Clin North Am 2002, 28:779–798. 18 Lundberg I, Brengman JM, Engel AG: Analysis of cytokine expression in muscle in inflammatory myopathies, Duchenne dystrophy, and non-weak controls. J Neuroimmunol 1995, 63:9–16. 19 Arahata K, Ishihara T, Fukunaga H, et al.: Inflammatory response in facioscapulohumeral muscular dystrophy (FSHD): immunocytochemical and genetic analyses. Muscle Nerve 1995, 2:S56–S66. 20 Fanin M, Angelini C: Muscle pathology in dysferlin deficiency. Neuropathol Appl Neurobiol 2002, 28:461–470. 21 Pegoraro E, Mancias P, Swerdlow SH, et al.: Congenital muscular dystrophy with primary laminin alpha2 (merosin) deficiency presenting as inflammatory myopathy. Ann Neurol 1996, 40:782–791. 22 Matsubara S, Hirai S, Sawa Y: Pulsed intravenous methylprednisolone therapy for inflammatory myopathies: evaluation of the effect by comparing two consecutive biopsies from the same muscle. J Neuroimmunol 1997, 76:75–80. 23 van der Pas J, Hengstman GJ, Ter Laak HJ, et al.: Diagnostic value of MHC class I staining in idiopathic inflammatory myopathies. J Neurol Neurosurg Psychiatry 2004, 75:136–139. 24 Civatte M, Schleinitz N, Krammer P, et al.: Class I MHC detection as a diagnostic tool in noninformative muscle biopsies of patients suffering from dermatomyositis (DM). Neuropathol Appl Neurobiol 2003, 29:546–552. Confalonieri P, Oliva L, Andreetta F, et al.: Muscle inflammation and MHC class I up-regulation in muscular dystrophy with lack of dysferlin: an immunopathological study. J Neuroimmunol 2003, 142:130–136. This is an interesting pathologic study comparing biopsy findings in dysferlinopathy with those seen in PM. 25 • 26 Nagaraju K, Raben N, Loeffler L, et al.: Conditional up-regulation of MHC class I in skeletal muscle leads to self-sustaining autoimmune myositis and myositis-specific autoantibodies. Proc Natl Acad Sci U S A 2000, 97:9209– 9214. 27 Bushby KM: Diagnostic criteria for the limb-girdle muscular dystrophies: report of the ENMC Consortium on limb-girdle dystrophies. Neuromuscul Disord 1995, 5:71–74. Kirschner J, Bonnemann CG: The congenital and limb-girdle muscular dystrophies: sharpening the focus, blurring the boundaries. Arch Neurol 2004, 61:189–199. This is a good general summary of the clinical and molecular aspects of congenital muscular dystrophies and LGMD. 28 • Is it really myositis? Nirmalananthan et al. 691 29 Zatz M, de Paula F, Starling A, et al.: The 10 autosomal recessive limb-girdle muscular dystrophies. Neuromuscul Disord 2003, 13:532–544. •• This is a detailed review of the clinical and molecular aspects of the recessive LGMDs. chenne muscular dystrophy and Becker muscular dystrophy among carriers in The Netherlands: a cohort study. Lancet 1999, 353:2116–2119. 44 Tupler R, Gabellini D: Molecular basis of facioscapulohumeral muscular dystrophy. Cell Mol Life Sci 2004, 61:557–566. 45 Kazemi-Esfarjani P, Skomorowska E, Jensen TD, et al.: A nonischemic forearm exercise test for McArdle disease. Ann Neurol 2002, 52:153–159. 46 Argov Z, Barash V, Soffer D, et al.: Late-onset muscular weakness in phosphofructokinase deficiency due to exon 5/intron 5 junction point mutation: a unique disorder or the natural course of this glycolytic disorder? Neurology 1994, 44:1097–1100. 47 Davis DB, Doherty KR, Delmonte AJ, et al.: Calcium-sensitive phospholipid binding properties of normal and mutant ferlin C2 domains. J Biol Chem 2002, 277:22883–22888. Sivakumar K, Vasconcelos O, Goldfarb L, et al.: Late-onset muscle weakness in partial phosphofructokinase deficiency: a unique myopathy with vacuoles, abnormal mitochondria, and absence of the common exon 5/intron 5 junction point mutation. Neurology 1996, 46:1337–1342. 48 Ausems MG, Lochman P, van Diggelen OP, et al.: A diagnostic protocol for adult-onset glycogen storage disease type II. Neurology 1999, 52:851–853. 34 Ueyama H, Kumamoto T, Horinouchi H, et al.: Clinical heterogeneity in dysferlinopathy. Intern Med 2002, 41:532–536. 49 Moufarrej NA, Bertorini TE: Respiratory insufficiency in adult-type acid maltase deficiency. South Med J 1993, 86:560–567. 35 Illarioshkin SN, Ivanova-Smolenskaya IA, Greenberg CR, et al.: Identical dysferlin mutation in limb-girdle muscular dystrophy type 2B and distal myopathy. Neurology 2000, 55:1931–1933. 50 Bornemann A, Besser R, Shin YS, et al.: A mild adult myopathic variant of type IV glycogenosis. Neuromuscul Disord 1996, 6:95–99. 51 Karmaniolas K, Ioannidis P, Liatis S, et al.: Primary carnitine deficiency in a male adult. J Med 2002, 33:105–110. 52 McFarland R, Taylor RW, Turnbull DM: The neurology of mitochondrial DNA disease. Lancet Neurol 2002, 1:343–351. 30 Hoffman EP, Rao D, Pachman LM: Clarifying the boundaries between the inflammatory and dystrophic myopathies: insights from molecular diagnostics and microarrays. Rheum Dis Clin North Am 2002, 28:743–757. 31 Stubgen JP: Limb girdle muscular dystrophy: a radiologic and manometric study of the pharynx and esophagus. Dysphagia 1996, 11:25–29. 32 Bansal D, Miyake K, Vogel SS, et al.: Defective membrane repair in dysferlindeficient muscular dystrophy. Nature 2003, 423:168–172. •• This excellent study sheds light on the biologic role of dysferlin in calciumdependent membrane fusion and repair. 33 36 McNally EM, Ly CT, Rosenmann H, et al.: Splicing mutation in dysferlin produces limb-girdle muscular dystrophy with inflammation. Am J Med Genet 2000, 91:305–312. 37 Rowin J, Meriggioli MN, Cochran EJ, et al.: Prominent inflammatory changes on muscle biopsy in patients with Miyoshi myopathy. Neuromuscul Disord 1999, 9:417–420. 38 Rosenberg NL, Ringel SP, Kotzin BL: Experimental autoimmune myositis in SJL/J mice. Clin Exp Immunol 1987, 68:117–129. 39 Bittner RE, Anderson LV, Burkhardt E, et al.: Dysferlin deletion in SJL mice (SJL-Dysf) defines a natural model for limb girdle muscular dystrophy 2B. Nat Genet 1999, 23:141–142. 40 Muntoni F, Torelli S, Ferlini A: Dystrophin and mutations: one gene, several proteins, multiple phenotypes. Lancet Neurol 2003, 2:731–740. •• This is an excellent review of the molecular genetics and associated phenotypes of the dystrophinopathies. 41 Escolar DM, Scacheri CG: Pharmacologic and genetic therapy for childhood muscular dystrophies. Curr Neurol Neurosci Rep 2001, 1:168–174. 42 Pegoraro E, Schimke RN, Garcia C, et al.: Genetic and biochemical normalization in female carriers of Duchenne muscular dystrophy: evidence for failure of dystrophin production in dystrophin-competent myonuclei. Neurology 1995, 45:677–690. 43 Hoogerwaard EM, Bakker E, Ippel PF, et al.: Signs and symptoms of Du- 53 Taylor RW, Schaefer AM, Barron MJ, et al.: The diagnosis of mitochondrial muscle disease. Neuromuscul Disord 2004, 14:237–245. • This review provides a useful diagnostic approach to mitochondrial myopathy. 54 Alshekhlee A, Kaminski HJ, Ruff RL: Neuromuscular manifestations of endocrine disorders. Neurol Clin 2002, 20:35–58, v–vi 55 Madariaga MG: Polymyositis-like syndrome in hypothyroidism: review of cases reported over the past twenty-five years. Thyroid 2002, 12:331–336. 56 Nishino I, Noguchi S, Murayama K, et al.: Distal myopathy with rimmed vacuoles is allelic to hereditary inclusion body myopathy. Neurology 2002, 59:1689–1693. 57 Yabe I, Higashi T, Kikuchi S, et al.: GNE mutations causing distal myopathy with rimmed vacuoles with inflammation. Neurology 2003, 61:384–386. • This case report demonstrates that patients with hereditary IBM may have inflammation on biopsy. 58 Fidzianska A, Rowinska-Marcinska K, Hausmanowa-Petrusewicz I: Coexistence of X-linked recessive Emery-Dreifuss muscular dystrophy with inclusion body myositis-like morphology. Acta Neuropathol (Berl) 2004, 107:197– 203. Myositis specific autoantibodies: changing insights in pathophysiology and clinical associations Gerald J.D. Hengstmana, Baziel G.M. van Engelena and Walther J. van Venrooijb Purpose of review Defined autoantibodies are found in about half of the patients with myositis. Traditionally, these autoantibodies have been divided into myositis specific autoantibodies (MSAs) and myositis associated autoantibodies. Several studies have shown that MSAs are associated with specific clinical characteristics and can aid our understanding of the pathophysiology of myositis. Recent findings Recent studies suggest that some MSAs are markers of specific inflammatory muscle diseases (e.g., anti-SRP for an immune-mediated necrotizing myopathy) and not just of myositis in general. Furthermore, new insights are emerging about the pathophysiology of MSAs, in particular anti-Jo-1. Based on these new insights, an alternative hypothesis of the formation of anti-Jo-1 autoantibodies is presented in which the immune system itself rather than muscle is the site of antigen presentation. Summary The recognition that some MSAs are markers of specific disease entities that were once commonly referred to as (poly)myositis, aids the development of better disease definitions. The changing insights in the function of the Jo-1 antigen and the emergence of new hypotheses on the formation of the Jo-1 antibody, open new avenues for future research aimed at unraveling the mystery of myositis. Keywords myositis, antibodies, autoantibodies, pathophysiology, differential diagnosis Curr Opin Rheumatol16:692–699. © 2004 Lippincott Williams & Wilkins. a Neuromuscular Centre Nijmegen, Department of Neurology, University Medical Centre Nijmegen, Nijmegen, The Netherlands and bNijmegen Centre for Molecular Life Sciences, Department of Biochemistry, University of Nijmegen, Nijmegen, The Netherlands This work was supported by NWO-MW, grant 940-37-009. Correspondence to Gerald J.D. Hengstman, Neuromuscular Centre Nijmegen, Department of Neurology, University Medical Centre Nijmegen, Internal mail number 326, PO Box 9101, 6500 HB Nijmegen, The Netherlands Tel: +31 24 3613396; fax: +31 24 3541122; e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:692–699 © 2004 Lippincott Williams & Wilkins 1040–8711 692 Introduction Autoantibodies can be found in the sera of most patients with myositis [1]. Defined autoantibodies are detected in about 50% of myositis patients and are traditionally divided into myositis specific autoantibodies (MSAs) and myositis associated autoantibodies (MAAs), the latter also occurring in autoimmune diseases without the presence of myositis [2]. Most MSAs are directed against cytoplasmic RNA-protein complexes involved in the process of protein synthesis [3–11]. The best-characterized MSAs are directed to several tRNA-synthetases and their cognate tRNAs, to components of the signal recognition particle (SRP), and to components of a nucleosome remodeling complex called Mi-2 [3–11]. Several clinical and epidemiologic studies have shown that MSAs are associated with specific clinical characteristics [12,13]. Table 1 gives an overview of the most common MSAs, their antigens, their frequency of occurrence in myositis patients, and their clinical associations. In the past couple of years, several studies have changed our insights about the MSAs. It is now becoming clear that some MSAs, in particular anti-Jo-1 and anti-signal recognition particle (anti-SRP) antibodies, are markers of very specific disease entities and not just of myositis in general. Furthermore, new ideas are emerging about the pathophysiology of MSAs, in particular anti-Jo-1. Changing diagnostic criteria Many studies have described the association of MSAs with specific subtypes of myositis and several extramuscular complications. Essential to the validity of these observed associations are the definitions used for diagnosis and identification of clinical signs and symptoms. Traditionally, most of the articles describing the MSA associations have used the Bohan and Peter criteria [14]. However, recent papers, especially in the neurologic literature, have challenged the concept of polymyositis (PM) [15–17]. The advocates for a more detailed histologic definition of PM state that inflammatory infiltrates in skeletal muscle tissue are not only present in PM but also in other myopathies including facioscapulohumeral dystrophy (FSHD), dysferlinopathies, etc. (Table 2) [15,16]. Bohan and Peter excluded only some of these diseases [14]. With present knowledge, it is probably best to state that all diseases mentioned in Table 2 should be excluded (on clinical grounds or histologically) MSAs: changing insights Hengstman et al. 693 Table 1. Overview of most common myositis specific autoantibodies Antibody Anti-ARS Anti-Jo-1 Anti-PL-7 Anti-PL-12 Anti-EJ Anti-OJ Anti-KS Anti-tRNA Anti-tRNAhis Anti-tRNAala Miscellaneous Anti-SRP Anti-Mi-2 Antigen His-tRNA synthetase Thr-tRNA synthetase Ala-tRNA synthetase Gly-tRNA synthetase Ile-tRNA synthetase Asp-tRNA synthetase tRNAhis tRNAala SRP-complex Nuclear helicase Frequency Clinical association 11–20% 2% 1% 1–3% 1% <1% Anti-synthetase syndrome Anti-synthetase syndrome Anti-synthetase syndrome Anti-synthetase syndrome Anti-synthetase syndrome Anti-synthetase syndrome 7% 1% Anti-synthetase syndrome Anti-synthetase syndrome 4% 4–14% “Aggressive” PM Classic DM Anti-ARS, anti-aminoacyl-tRNA synthatase; SRP, signal recognition particle; PM, polymyositis; DM, dermatomyositis. in patients suspected of having PM. It is because of these ongoing changes in diagnostic criteria that associations we thought were once clear (eg, anti-SRP is only seen in PM) are now being reconsidered (eg, anti-SRP is specific for a type of necrotizing myopathy). Anti-aminoacyl-tRNA synthetases The most prevalent MSAs are directed against aminoacyl-tRNA synthetases (ARS). The presence of anti-ARS antibodies is strongly associated with the anti-synthetase syndrome, consisting of myositis, idiopathic interstitial lung disease (ILD), nonerosive arthritis, and Raynaud’s phenomenon [12,18–21]. Anti-Jo-1 and interstitial lung disease A recent prospective study performed by Fathi et al. [22] confirmed the association of anti-Jo-1 with ILD and arthritis. A small group of 17 newly diagnosed dermatomyositis (DM)/PM patients were investigated for the presence of ILD defined as the occurrence of radiographic signs of ILD on chest X-ray or high-resolution CT scan (HRCT) and/or restrictive ventilatory defect. Eleven patients were diagnosed with ILD, four of whom had the anti-Jo-1 autoantibody. Of the six patients without ILD, none was anti-Jo-1 positive. Another retrospective study also examined the presence of ILD in DM/PM and compared ILD in patients with Table 2. Myopathies with inflammatory infiltrates on muscle biopsy Immune-mediated myopathies Dermatomyositis Inclusion body myositis Polymyositis Muscular dystrophies Duchenne muscular dystrophy Becker muscular dystrophy Facioscapulohumeral muscular dystrophy (FSHD) Dysferlinopathies Limb-girdle muscular dystrophy Necrotizing myopathies Toxic myopathies and without anti-Jo-1 [23]. In a group of 156 patients, ILD was diagnosed in 23.1%. The anti-Jo-1 autoantibody was found in only 15 patients (9%), a remarkably low number of patients raising some questions on the methodology of the study, especially since the authors do not describe the serological tests used. The prevalence of ILD in the anti-Jo-1 group was 73%, again confirming the association of this antibody with ILD. The only differences found between the anti-Jo-1 positive and anti-Jo-1 negative group were that the anti-Jo-1 positive group were less likely to have a symptomatic form of ILD and bronchiolitis obliterans organizing pneumonia (BOOP). The authors noticed that patients with antiJo-1 had similar ILD outcome, compared with those without this antibody, with respect to resolution, improvement, or deterioration of ILD and mortality rate related to ILD complications. They concluded that patients with and without anti-Jo-1 require similar management and follow-up of ILD. Specificity of anti-Jo-1 Although it has been shown that anti-Jo-1 is specific for DM/PM compared with other inflammatory autoimmune rheumatic disorders, few studies have investigated its specificity compared with other neuromuscular disorders [19,24–26]. In a small study of 17 patients with Duchenne muscular dystrophy, anti-Jo-1 was not found [25]. In another study, Tanimoto et al. [26] were unable to detect the anti-Jo-1 autoantibody in sera from 33 patients with a neuromuscular disorder other than myositis. Unfortunately, they did not describe their serological technique nor specify the type of neuromuscular disorders examined. In a recent study, we were unable to detect anti-Jo-1 in sera from 18 patients with FSHD, a muscular dystrophy with marked inflammation on muscle biopsy [27]. Based on these findings, it can be concluded that the anti-Jo-1 autoantibody is highly specific for DM/PM and that the formation of anti-Jo-1 is not merely the result of muscle inflammation but is closely linked to the pathophysiology of DM/PM. 694 Myositis and myopathies The anti-synthetase syndrome Because earlier studies only looked for anti-ARS antibodies in patients with myositis and other autoimmune rheumatic tissue diseases, and only found them in myositis patients, it was thought that anti-ARS antibodies are myositis specific. Based on more recent work, it became clear that anti-ARS antibodies are specific for their own disease entity of which myositis may be a component: the anti-synthetase syndrome (Table 3) [28,29]. Histologic studies confirmed that the anti-synthetase syndrome is a separate disease entity within the spectrum of myositis [30]. Mozaffar and Pestronk [30] demonstrated that the myopathological changes in the anti-synthetase syndrome include perimysial connective tissue fragmentation and inflammation, with muscle fiber pathology in neighboring perifascicular regions. Based on their findings they suggested that myositis in the anti-synthetase syndrome may result from an immune-mediated disorder of connective tissue. This hypothesis can explain more easily why ILD is such a prominent feature of the antisynthetase syndrome. Kamei [35•] examined the cellular localization of Jo-1 tagged with green fluorescent protein in transfected T24 cells. The tagged-Jo-1 localized solely and diffusely in the cytoplasm of almost all cells. Occasionally though, small aggregates or spots could be seen in or near the nucleus. The author subsequently demonstrated that these seemingly nuclear spots coincide with cytoplasmic invaginations of the nuclear membrane, indicating that tagged-Jo-1 is not localized in the nucleus but solely in the cytoplasm. Anti-Jo-1 antibody formation Ever since its first description almost 25 years ago, researchers have hoped that the anti-Jo-1 autoantibody would provide a better understanding of the pathogenesis of myositis. To elucidate the role of anti-Jo-1 in myositis, two questions must be answered: why are they formed and what do they do? Neither question can be fully answered but new insights are emerging based on recent studies casting a different light on the Jo-1 antigen. Earlier work has shown that the anti-Jo-1 autoantibody response is antigen-driven and very closely linked to the disease process [36]. There are several hypotheses as to why anti-Jo-1 is the subject of antibody targeting. One is that anti-Jo-1 autoantibodies result from a direct interaction of Jo-1 with RNA from picornaviruses, thus rendering it foreign to the immune system [6]. Another hypothesis proposes that the immune response is primarily directed against picornaviral proteins with regions homologous to regions present in Jo-1, thus causing autoantibody formation via the mechanism of molecular mimicry [37]. A third hypothesis is based on the formation of anti-idiotypic antibodies, again triggered by a presumed viral protein [38]. A more recent model hypothesizes that certain self-proteins become modified (e.g., during apoptosis or as a result of inflammatory or infectious processes) and are subsequently recognized as nonself by the immune system [39]. The immune response might then, via epitope spreading, evolve into a fullblown immune reaction directed to the whole protein, including the nonmodified parts. Cellular localization of Jo-1 Jo-1 and the immune system Knowledge of the cellular localization of an autoantigen is important for the understanding of its cellular function and for gaining further insight in the pathogenesis of the disease studied. Anti-Jo-1 autoantibodies are directed against His-tRNA-synthetase, an enzyme with a cytoplasmic function and therefore a presumably cytoplasmic localization. However, contradictory results have been reported regarding the cellular localization of Jo-1. Cytoplasmic, nuclear, nucleolar, and all forms of combinations of these localizations have been reported [1,31–34]. Jo-1 has traditionally been seen as a ubiquitously expressed intracellular protein catalyzing the binding between the amino acid histidine and its tRNA. But why is Jo-1 the target of a disease-specific antibody response? Because anti-Jo-1 antibodies were thought to occur only in patients with myositis, most studies have looked at the muscle as the site of antigen expression. This notion led to the development of a hypothesis in which a presumably altered Jo-1 is expressed by muscle tissue thus eliciting an immune reaction directed against muscle tissue along with a specific antibody response (Fig. 1A). Others have postulated that necrosis or apoptosis of muscle fibers causes modifications of Jo-1, which are subsequently exposed to the immune system as the muscle fibers disintegrate (Fig. 1B). Recent studies, however, showed that fragments of several tRNA synthetases function as chemokines with a potential role in the immune response of myositis [40,41••]. It is thus conceivable that anti-Jo-1 antibodies are not directed to the entire Jo-1 antigen but initially only against its chemokinefragment. Through epitope-spreading, antibodies can eventually be formed against other components of Jo-1. Immunopathogenesis of the anti-Jo-1 autoantibody Table 3. Characteristics of the anti-synthetase syndrome Clinical Laboratory Biopsy Treatment Myositis Interstitial lung disease Nonerosive arthritis Raynaud phenomenon Anti-ARS antibodies Fragmentation of perimysial connective tissue Macrophage predominant perimysial inflammation Perifascicular myopathic changes Normal capillary density Normal response of interstital lung disease to treatment Moderate response of myositis to treatment MSAs: changing insights Hengstman et al. 695 Figure 1. Hypotheses on the role of the Jo-1 antigen and the formation of anti-Jo-1 autoantibodies in myositis (A) Initiated by a factor X (step 1), altered Jo-1 is formed (step 2) and presented by MHC class I molecules on the sarcolemma (step 3) to the immune system (step 4) with subsequent formation of anti-Jo-1 antibodies. (B) Inflammation of muscle tissue (step 1) results in necrosis/apoptosis of muscle cells. In this process of necrosis/apoptosis, Jo-1 is altered (posttranslational modification) (step 2). As the cell disintegrates, the altered Jo-1 antigen is phagocytized (step 3 and 4) and subsequently presented to the immune system by antigen-presenting cells (step 5), resulting in formation of anti-Jo-1 antibodies (step 6). (C) Initiated by factor X, immune cells form the chemokine-fragment of Jo-1 (step 1), which is subsequently secreted together with other pro-inflammatory molecules (step 2) resulting in inflammation of muscle tissue and induction of aberrant MHC class I expression on the sarcolemma (step 3). 696 Myositis and myopathies If the chemokine-fragment is the actual antigen, it becomes more likely that the immune system rather than muscle is the site where the initial antigen is expressed (Fig. 1C). The latter hypothesis is attractive for several reasons. First, one can now understand better why myositis is a multisystem disorder. If the muscle is the initial site of the immune response, it is hard to understand why the lung, or the joints, or the skin become involved. Several studies have shown that patients with the anti-Jo-1 autoantibody do not always have myositis at the time of the detection of the antibody [28,29]. Cases like these cannot be explained by anti-Jo-1 formation in muscle tissue. But why are antibodies formed against a chemokine? Several explanations are possible. First, the immune response is always a battlefield between proinflammatory and antiinflammatory components. As part of the antiinflammatory reaction, antibodies may be formed, which suppress the function of proinflammatory molecules, like chemokines. The formation of anti-Jo-1 antibodies may thus be a consequence of the immune system trying to suppress the immune response. Secondly, the immune response in myositis may be abnormal with formation of abnormal chemokines, which are foreign to the immune system. Thirdly, apoptosis of inflammatory cells plays an important role in the regulation of the immune response. Through abnormal apoptotic cleavage, unique fragments of Jo-1 may be formed and presented to the immune system, causing the formation of anti-Jo-1 antibodies. Jo-1–induced T-cell proliferation In an interesting study, Ascherman et al. [42] looked at antigen-presenting cells (APCs) and their role in eliciting a T-cell response to the Jo-1 antigen. Using recombinant full-length Jo-1 (generated from the RNA of a healthy control subject) and four fragments of this recombinant antigen, T-cells from the peripheral blood of anti-Jo-1– positive myositis patients and from healthy controls were stimulated in the presence of PBMC-derived APCs and dendritic cells (DCs). In anti-Jo-1–positive patients and in healthy controls T-cell proliferation was found to both full-length Jo-1 and to its individual fragments in a dosedependent manner. An absolute dependence on DCs was found for the productive presentation of Jo-1 fragments, and this was not the case for the productive presentation of full-length Jo-1. It was further shown, through antibody-blocking experiments, that the T-cell proliferation driven by full-length Jo-1 as well as by Jo-1 fragments is MHC class II dependent. Although the authors demonstrated elegantly that Jo-1 and its fragments can induce a T-cell proliferation, they failed to identify differences between healthy controls and anti-Jo-1– positive patients. A potential explanation for this is that they used a recombinant Jo-1 product that does not con- tain protein modifications. Based on the hypothesis that Jo-1 might be altered in anti-Jo-1 positive myositis patients (eg, through posttranslational modifications), it would have been of interest to see whether their experiments would have rendered different results if they had used Jo-1 isolated from biopsies of anti-Jo-1–positive patients. Anti-signal recognition particle Several studies have shown that anti-signal recognition particle (anti-SRP) autoantibodies are mainly found in PM (defined by the Bohan and Peter criteria), and only occasionally in DM and inclusion body myositis (IBM) [2,8,12,14,18,19,43–46]. Although few patients were studied, an association between the antibodies and an acute and severe myositis, cardiac involvement, a poor response to treatment, and an increased mortality rate [12,18,19,44] was noted. As we have pointed out previously, many of these presumed associations of anti-SRP autoantibodies are weak [39]. A recent study confirmed the presence of a relatively aggressive disease characterized by severe myalgia and arthralgia, high levels of serum creatine kinase, and a moderate response to immunosuppressive treatment in patients with the anti-SRP antibody [43]. Cardiac involvement, however, was not found [43]. Anti-signal recognition particle myopathy Recent studies have shown that anti-SRP autoantibodies are markers for a specific immune-mediated myopathy other than PM. In an excellent paper Miller et al. [47] described the clinical and pathologic features of seven patients with anti-SRP autoantibodies as detected by two different techniques: immunoprecipitation and immunodiffusion. The onset of weakness in all patients was between August and January. All patients progressed relatively rapidly to severe weakness with a mean time from onset to maximum weakness of 5 months. Most patients complained of fatigue or pain, but this was never the primary complaint. Physical examination revealed a severe symmetric proximal weakness of the upper and lower extremities. None of the patients had clinical signs or symptoms suggestive of cardiac involvement. Serum creatine kinase levels were markedly elevated with a mean of 12.900 U/l and electromyography showed myopathic features with very prominent spontaneous activity. Six of the seven patients improved on corticosteroids, but only partially. Three patients relapsed after tapering of the corticosteroids. The histopathology in the seven patients was remarkable. All biopsies showed myopathic features with regenerating fibers. Mononuclear inflammatory infiltrates were uncommon and there was no marked MHC-I staining of the sarcolemma. Necrotic muscle fibers and increased endomysial connective tissue were noticed in MSAs: changing insights Hengstman et al. 697 Table 4. Characteristics of the anti-SRP myopathy Clinical Laboratory EMG Biopsy Treatment Rapidly progressive disease Severe weakness Symmetric proximal weakness of arms and legs Marked atrophy of proximal muscles High levels of serum CK Marked spontaneous activity on EMG Absence of inflammatory infiltrates Absence of HLA-ABC class antigens on the sarcolemma Responsive to treatment, but only partial most biopsies as well as a reduced endomysial capillary density. Furthermore, the mean diameter of the endomysial capillaries was increased with deposition of membrane attack complex (MAC). The histopathology Miller et al. found in their anti-SRP patients strongly differs from the histopathology of PM in which mononuclear inflammatory infiltrates and MHC-class I staining of the sarcolemma are usually present, the diameter of endomysial capillaries is not altered, and deposition of MAC in the capillaries is absent [30,47,48]. Miller et al. thus concluded that the anti-SRP autoantibodies identify a specific immune- mediated myopathy characterized by a rapidly progressive severe proximal muscle weakness with an incomplete response to corticosteroids and no clinical signs or symptoms suggestive of multi-organ involvement. Based on the histopathological features, the authors hypothesized that the disease referred to as “myopathy with anti-SRP autoantibodies” is caused by a humoral immune mechanism primarily directed against the endomysial capillaries with subsequent multifocal ischemic pathology of the muscle tissue. Where the formation of antibodies directed against components of the SRP complex fits into the picture remains unknown. Second clinical study on anti- signal recognition particle myopathy In another study, Kao et al. [49•] determined the longterm outcome and associated clinical, serological, and pathologic features of 19 patients with the anti-SRP autoantibody. Sera from 263 patients with DM/PM, 790 patients with systemic sclerosis (SSc), and 109 patients with an overlap syndrome were examined. Sixteen sera tested positive for anti-SRP in the myositis group, two in the SSc group, and one in the overlap syndrome group. All patients in the myositis group were diagnosed with PM according to the Bohan and Peter criteria [14]. The authors subsequently compared the anti-SRP positive PM patients with the anti-SRP negative PM patients. No differences were observed with regard to demographic features (no clear seasonal onset, although the authors do not state how they identified the month of onset), cardiac involvement (defined as echocardiographic evidence of biventricular cardiomyopathy or electrocardiographic evidence of ventricular arrhythmias attributable to PM and not to coronary heart disease), or 5-year cumulative survival rate (86%). The patients with anti-SRP autoantibodies had more frequently severe proximal weakness at presentation, higher levels of serum CK at diagnosis, and more severe muscle atrophy at presentation. Three anti-SRP–positive PM patients were diagnosed with ILD and two with arthritis. The authors noticed that the anti-SRP–positive PM patients exhibited persistent muscle weakness and resistance to treatment, with a favorable response being achieved in only one third. Unfortunately, no clear data are presented on which these conclusions are based. The muscle biopsy specimens of 10 PM patients with anti-SRP autoantibodies were compared with those of 17 PM patients without the antibody. Compatible with the findings by Miller et al., the authors Figure 2. Different disease entities within the spectrum of myositis, past and present More than 25 years ago, myositis was roughly divided into DM and PM. With the present day knowledge, we are able to identify several distinct disease entities that were formerly included in dermatomyositis (DM) and polymyositis (PM). Several of these disease entities are characterized by certain specific autoantibodies, as indicated. anti-SRP, anti-signal recognition particle antibodies; anti-ARS, anti-aminoacyl-tRNA synthetase antibodies. 698 Myositis and myopathies did not find extensive endomysial inflammation, but unlike Miller et al. they also did not notice marked necrosis [47]. The authors state that the two patients with SSc and the one patient with an overlap syndrome (suffering from the anti-synthetase syndrome) had no features of myositis, without mentioning what they mean with “features” (are these clinical signs and symptoms, or an elevated serum CK or muscle biopsy abnormalities?). Based on their study, the authors conclude that anti-SRP is not specific for PM, that severe muscle weakness and atrophy are prominent features whereas cardiac involvement is less common, and survival is better than previously reported. In conclusion, the contours of a specific myopathy are emerging that can be identified by the presence of antiSRP autoantibodies (Table 4). However, several questions remain unanswered including the presence of necrotic fibers and the predilection of the disease to start in the fall. Conclusion In conclusion, the MSAs have aided in the identification of several distinct disease entities within the spectrum of myositis. More than 25 years ago, PM was defined as an acquired muscle weakness with the presence of inflammatory infiltrates in skeletal muscle, usually responsive to immunosuppression. Today we recognize several different diseases that once were included under the heading PM (Fig. 2). Besides histology and accurate clinical descriptions, MSAs have played an important role in this process. As several authors have pointed out, PM is most frequently a component of an overlap syndrome and with current knowledge, we are able to diagnose most of these patients correctly [15,16]. These changes in disease definitions will aid research in unraveling the pathogenesis of these diseases and in performing better therapeutic trials. Furthermore, the increasing insights in the function of the Jo-1 antigen and the emergence of new hypotheses on the formation of the anti-Jo-1 antibody, open new avenues for future research aimed at unraveling the mystery of these disabling diseases. 4 Bunn CC, Bernstein RM, Mathews MB: Autoantibodies against alanyl-tRNA synthetase and tRNAala coexist and are associated with myositis. J Exp Med 1986, 163:1281–1291. 5 Hirakata M, Suwa A, Nagai S, et al.: Anti-KS: identification of autoantibodies to asparaginyl-transfer RNA synthetase associated with interstitial lung disease. J Immunol 1999, 162:2315–2320. 6 Mathews MB, Bernstein RM: Myositis autoantibody inhibits histidyl-tRNA synthetase: a model for autoimmunity. Nature 1983, 304:177–179. 7 Mathews MB, Reichlin M, Hughes GRV, et al.: Anti-threonyl-tRNA synthetase, a second myositis-related autoantibody. J Exp Med 1984, 160:420–434. 8 Reeves WH, Nigam SK, Blobel G: Human autoantibodies reactive with the signal-recognition particle. Proc Natl Acad Sci USA 1986, 83:9507–9511. 9 Targoff IN, Reichlin M: The association between Mi-2 antibodies and dermatomyositis. Arthritis Rheum 1985, 28:796–803. 10 Targoff IN: Autoantibodies to aminoacyl-transfer RNA synthetases for isoleucine and glycine. Two additional synthetases are antigenic in myositis. J Immunol 1990, 144:1737–1743. 11 Zhang Y, LeRoy G, Seelig HP, et al.: The dermatomyositis-specific autoantigen Mi2 is a component of a complex containing histone deacetylase and nucleosome remodeling activities. Cell 1998, 95:279–289. 12 Love LA, Leff RL, Fraser DD, et al.: A new approach to the classification of idiopathic inflammatory myopathy: myositis-specific autoantibodies define useful homogeneous patient groups. Medicine 1991, 70:360–374. 13 Joffe MM, Love LA, Leff RL, et al.: Drug therapy of the idiopathic inflammatory myopathies: predictors of response to prednisone, azathioprine, and methotrexate and a comparison of their efficacy. Am J Med 1993, 94:379–387. 14 Bohan A, Peter JB: Polymyositis and dermatomyositis (first of two parts). N Engl J Med 1975, 292:344–347. 15 Dalakas MC, Hohlfeld R: Polymyositis and dermatomyositis. Lancet 2003, 362:971–982. 16 Amato AA, Griggs RC: Unicorns, dragons, polymyositis, and other mythological beasts. Neurology 2003, 61:288–290. 17 Van der Meulen MFG, Bronner IM, Hoogendijk JE, et al.: Polymyositis, an overdiagnosed entity. Neurology 2003, 61:316–321. 18 Hausmanowa-Petrusewicz I, Kowalska-Oledzka E, Miller F, et al.: Clinical, serological, and immunogenetic features of Polish patients with idiopathic inflammatory myopathies. Arthritis Rheum 1997, 40:1257–1266. 19 Hirakata M, Mimori T, Akizuki M, et al.: Autoantibodies to small nuclear and cytoplasmic ribonucleoproteins in Japanese patients with inflammatory muscle disease. Arthritis Rheum 1992, 35:449–456. 20 Mimori T: Structures targeted by the immune system in myositis. Curr Opin Rheumatol 1996, 8:521–527. 21 Targoff IN: Immune manifestations in inflammatory muscle disease. Rheum Dis Clin North Am 1994, 20:857–880. 22 Fathi M, Dastmalchi M, Rasmussen E, et al.: Interstitial lung disease, a common manifestation of newly diagnosed polymyositis and dermatomyositis. Ann Rheum Dis 2004, 63:297–301. 23 Marie I, Hachulla E, Chérin P, et al.: Interstitial lung disease in polymyositis and dermatomyositis. Arthritis Rheum 2002, 47:614–622. 24 Vázquez-Abad D, Rothfield NF: Sensitivity and specificity of anti-Jo-1 antibodies in autoimmune disease with myositis. Arthritis Rheum 1996, 39:292–296. 25 Perurena O, Targoff IN, Bobele G, et al.: Lack of myositis-specific autoantibodies in Duchenne’s muscular dystrophy. Ann Neurol 1993, 34:308. 26 Tanimoto K, Nakano K, Kano S, et al.: Classification criteria for polymyositis and dermatomyositis. J Rheumatol 1995, 22:666–674. 27 Hengstman GJD, Van der Kooi EL, Vree Egberts WTM, et al.: Anti-Jo-1 autoantibodies aid in differentiating facioscapulohumeral dystrophy from polymyositis. Neurology 2004, 62(suppl 5):A500. 28 Friedman AW, Targoff IN, Arnett FC: Interstitial lung disease with autoantibodies against aminoacyl-tRNA synthetases in the absence of clinically apparent myositis. Semin Arthritis Rheum 1996, 26:459–467. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest 1 Reichlin M, Arnett FC: Multiplicity of antibodies in myositis sera. Arthritis Rheum 1984, 27:1150–1156. 29 2 Brouwer R, Hengstman GJD, Vree Egberts W, et al.: Autoantibody profiles in the sera of European patients with myositis. Ann Rheum Dis 2001, 60:116– 123. Schmidt WA, Wetzel W, Friedländer R, et al.: Clinical and serological aspects of patients with anti-Jo-1 autoantibodies—an evolving spectrum of disease manifestations. Clin Rheumatol 2000, 19:371–377. 30 3 Brouwer R, Vree Egberts W, Jongen PH, et al.: Frequent occurrence of antitRNAhis autoantibodies that recognize a conformational epitope in sera of patients with myositis. Arthritis Rheum 1998, 41:1428–1437. Mozaffar T, Pestronk A: Myopathy with anti-Jo-1 antibodies: pathology in perimysium and neighbouring muscle fibres. J Neurol Neurosurg Psychiatry 2000, 68:472–478. 31 Nishikai M, Reichlin M: Heterogeneity of precipitating antibodies in polymyo- MSAs: changing insights Hengstman et al. 699 sitis and dermatomyositis. Characterization of the Jo-1 antibody system. Arthritis Rheum 1980, 23:881–888. 32 Rosa MD, Hendrick JP Jr, Lerner MR, et al.: A mammalian tRNAHis-containing antigen is recognized by the polymyositis-specific antibody anti-Jo-1. Nucleic Acids Res 1983, 11:853–870. 33 Shi MH, Tsui FWL, Rubin LA: Cellular localization of the target structures recognized by the anti-Jo-1 antibody: immunofluoresence studies on cultured human myoblasts. J Rheumatol 1991, 18:252–258. 34 Vázquez-Abad D, Carson JH, Rothfield N: Localization of histidyl-tRNA synthetase (Jo-1) in human laryngeal epithelial carcinoma cell line (HEp-2 cells). Cell Tissue Res 1996, 286:487–491. 35 Kamei H: intracellular localization of histidyl-tRNA synthetase/Jo-1 antigen in T24 cells and some other cells. J Autoimmun 2004, 22:201–210. • In a detailed study it is shown that the Jo-1 antigen (His-tRNA synthetase) is exclusively localized in the cytoplasm. 36 37 Miller FW, Twitty SA, Biswas T, Plotz PH: Origin and regulation of a diseasespecific autoantibody response. Antigenic epitopes, spectrotype stability, and isotype restriction of anti-Jo-1 autoantibodies. J Clin Invest 1990, 85:468–475. Walker EJ, Jeffrey PD: Sequence homology between encephalomyocarditis virus protein VPI and histidyl-tRNA synthetase supports a hypothesis of molecular mimicry in polymyositis. Med Hypotheses 1988, 25:21–25. 38 Plotz PH: Autoantibodies are anti-idiotype antibodies to antiviral antibodies. Lancet 1983, 2:824–826. 39 Hengstman GJD, Van Engelen BGM, Vree Egberts WTM, Van Venrooij WJ: Myositis-specific autoantibodies: overview and recent developments. Curr Opin Rheumatol 2001, 13:476–482. 40 Wakasugi K, Schimmel P: Highly differentiated motifs responsible for two cytokine activities of a split human tRNA synthetase. J Biol Chem 1999, 274:23155–23159. 41 •• Howard OMZ, Dong HF, Yang D, et al.: Histidyl-tRNA synthetase and asparaginyl-tRNA synthetase, autoantigens in myositis, activate chemokine recep- tors on T lymphocytes and immature dendritic cells. J Exp Med 2002, 196:781–791. Two well-characterized autoantigens in myositis are found to be chemokines, thus adding a new dimension to the function of these autoantigens. 42 Ascherman DP, Oriss TB, Oddis CV, Wright TM: Critical requirement for professional APCs in eliciting T cell responses to novel fragments of histidyltRNA synthetase (Jo-1) in Jo-1 antibody-positive polymyositis. J Immunol 2002, 169:7127–7134. 43 Hengstman GJD, Brouwer R, Vree Egberts WTM, et al.: Clinical and serological characteristics of 125 Dutch myositis patients; myositis specific autoantibodies aid in the differential diagnosis of the idiopathic inflammatory myopathies. J Neurol 2002, 249:69–75. 44 Targoff IN, Johnson AE, Miller FW: Antibody to signal recognition particle. Arthritis Rheum 1990, 33:1361–1370. 45 Arnett FC, Targoff IN, Mimori T, et al.: Interrelationship of major histocompatibility complex class II alleles and autoantibodies in four ethnic groups with various forms of myositis. Arthritis Rheum 1996, 39:1507–1518. 46 Hirakata M, Nakamura K, Fuji J, et al.: Clinical and immunogenetic features of anti-SRP autoantibodies in Japanese patients. Arthritis Rheum 1995, 38:S321. 47 Miller T, Al-Lozi MT, Lopate G, Pestronk A: Myopathy with antibodies to the signal recognition particle: clinical and pathological features. J Neurol Neurosurg Psychiatry 2002, 73:420–428. 48 Van der Pas J, Hengstman GJD, Ter Laak HJ, Van Engelen BGM: Diagnostic value of MHC class I staining in idiopathic inflammatory myopathies. J Neurol Neurosurg Psychiatry 2004, 75:136–139. Kao AH, Lacomis D, Lucas M, et al.: Anti-signal recognition particle autoantibody in patients with and patients without idiopathic inflammatory myopathy. Arthritis Rheum 2004, 50:209–215. Several clinical associations of anti-SRP autoantibodies are confirmed, thus strengthening the observation that this antibody is associated with its own specific disease. It is further shown that some patients with anti-SRP have extramuscular involvement. 49 • Myositis: an update on pathogenesis Lisa Christopher-Stinea and Paul H. Plotzb Purpose of review The etiology and much about the pathogenesis of the inflammatory myopathies remain a mystery. In this review, we investigate recent research efforts to understand the pathogenesis of the diverse entities of polymyositis (PM), dermatomyositis (DM), and inclusion body myositis (IBM), diseases that result from interactions between environmental risk factors and genetic susceptibility. Recent findings Over the past year, there has been considerable progress toward better understanding of IBM, with relatively few developments toward understanding PM and DM. Although these diseases may share some common clinical phenotypic and serologic components, they differ on a molecular and cellular level. Summary The need for definitive, safer therapies in these diseases makes vital the search for defining detailed pathogenesis of inflammation and muscle fiber damage at the molecular level. Keywords inflammatory myopathy, polymyositis, dermatomyositis, inclusion body myositis, pathogenesis Curr Opin Rheumatol16:700–706. © 2004 Lippincott Williams & Wilkins. a Instructor in Medicine, Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA and bChief, Arthritis and Rheumatism Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA Correspondence to Paul H. Plotz, Chief, Arthritis and Rheumatism Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Clinical Center 9N244, Bethesda, MD 20892-1820, USA Tel: 301 496 9904; fax: 301 402 0012; e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:700–706 © 2004 Lippincott Williams & Wilkins 1040–8711 700 Introduction The etiology and much about the pathogenesis of the inflammatory myopathies remain elusive. We review here recent scientific endeavors exploring the pathogenesis of the diverse entities of polymyositis (PM), dermatomyositis (DM), and inclusion body myositis (IBM), diseases that are held to result from interactions between environmental risk factors and genetic susceptibility. There have been relatively few developments toward understanding PM and DM pathogenesis in the past year, but considerable progress toward better understanding of IBM. Polymyositis and dermatomyositis Experimental models A Swedish study examined the skeletal muscle changes in an experimental model of Chagas disease and compared its muscle inflammation pattern with that of the human inflammatory myopathies [1••]. Female CBA/J mice were injected with the Tulahuen strain of T. cruzi, and the inflammatory phenotype was characterized. The predominant muscle cell types in the model were macrophages and T cells, with CD4+ cells near blood vessels and CD8+ cells present within the perimysium; almost no B cells were present; no vacuolar fibers were noted. Thus, the model most closely approximates PM. This may be a useful animal model for PM, particularly as it relates to understanding the order of events that lead to inflammation and muscle damage in association with Tcell infiltration. Clinical aspects Potential association with viral illness Attempts to find evidence of a direct role of viral infection in myositis have continued, though again without really credible success. A rare example of a viral myopathy seen in conjunction with the West Nile Virus was noted in one case report [2]. Symptoms in addition to the neurologic manifestations included a myositis characterized by a T-lymphocyte infiltration of nerve fibers, leading the authors to conclude that the virus may reach the central nervous system via peripheral nerves. There was muscle fiber necrosis, atrophy, and inflammation with an exclusive Tcell infiltration with a slight predominance of CD4+ over CD8+ cells and with scattered CD68+ cells. There were no inflammatory changes seen in blood vessels and muscle septa, and WNV antigens were not detected Myositis Christopher-Stine and Plotz 701 by immunohistochemistry. This appearance suggests that the myalgia often reported with a self-limited WNV illness [3] might be a viral myositis rather than evidence of neurologic involvement. Douche-Aourik et al. [4] reported the persistence of enterovirus RNA in muscle samples of patients with inflammatory myopathies and fibromyalgia. There was no evidence of VP-1 protein in the samples positive for putative viral RNA. These findings must be read with skepticism. The authors fail to cite the strongest evidence that enteroviral sequences are absent from myositis samples—a study in which the potential technical problems that they name, and others they missed, had been carefully anticipated and excluded [5]. A French [6] study was unable to detect B19 DNA by PCR in 7 of 8 muscle biopsy samples obtained from patients with inflammatory myopathy, and viral capsid protein expression was not demonstrated by immunohistochemistry. One patient had a transiently positive viral capsid protein that became negative even in the absence of disease flare. Parvovirus B19 was unable to induce IL-6 production by myoblasts in vitro. Interstitial lung disease Fathi et al. [7•] investigated the prevalence and disease predictors for ILD in 17 patients newly diagnosed with PM or DM in Sweden. Sixty-five percent had evidence of pulmonary involvement, even in the absence of symptoms. Arthritis and anti-Jo-1 antibodies were predictors of concomitant pulmonary involvement. We agree with the authors’ suggestion that chest x-ray, HRCT, PFTs, and anti-Jo-1 antibodies should be included in the initial investigation of PM and DM patients. Schnabel et al. [8•] screened 63 consecutive PM/DM patients for lung disease. They found evidence of ILD in 32% of subjects screened. Those patients who progressed were distinguished by evidence of ground-glass opacities on HRCT and by BAL neutrophilia. Cyclophosphamide provided stabilization, and sometimes improvement, in all members of this group. Those patients without rapidly progressive disease had stable lung disease on moderateintensity immunosuppressives during the mean of 35 months of follow-up. This study suggests that treatment for ILD in future clinical trials for PM/DM-associated lung disease should stratify patients. Nonspecific interstitial pneumonia was the most common observed histologic pattern in patients with clinically amyopathic dermatomyositis in a retrospective multicenter study performed in France [9]. A recent case report reminds us that fatal interstitial fibrosis may be present in the presence of clinically amyopathic dermatomyositis, even if there is no evidence of anti-Jo-1 or other antisynthetase antibodies [10]. Necrotizing myopathy On occasion, patients presenting with a clinically apparent idiopathic inflammatory myopathy may lack mononuclear cell infiltrate on muscle biopsy despite the presence of a necrotizing myopathy [11]. This subset of patients may still be steroid responsive, as evidenced by this report, thus suggesting that clinicians not delay steroid therapy despite lack of a monocellular infiltrate when the rest of the clinical picture suggests acute myositis. De Bleecker et al. [12•] report a case of a patient with an ill-defined overlap syndrome including a chronic-steroid responsive necrotizing myopathy in the absence of a malignancy. The muscle biopsy demonstrated little inflammation, but there was MAC deposition on the mural elements of the vessel wall, and MHC class I was present on the surface of all muscle fibers. Thus, virtual absence of inflammatory changes need not exclude the diagnosis of a potentially treatable autoimmune inflammatory myopathy. The pathogenesis in this interesting disease subset needs study. Environmental risk factors In the most interesting and persuasive evidence of an environmental contribution to the expression of an autoimmune rheumatic disease, Okada et al. [13••] demonstrated that among 13 geoclimatic variables studied in a population of 919 consecutive myositis patients from 15 locations surface ultraviolet radiation intensity was the strongest contributor to DM and was strongly related to the proportion of anti-Mi2 antibodies. These data suggest that ultraviolet radiation exposure may modulate the expression of autoimmune diseases such as myositis both immunologically and clinically in different populations around the world. This may be related to the observation that, at least in DM, increased numbers of ultraviolet light-induced apoptotic cells in the skin can lead to a supra-threshold concentration of antigenic peptides [14]. Cellular immune mechanisms Tumor necrosis factor-␣ Kuru et al. [15] studied the expression of TNF-␣ and its receptor in PM, DM, and Duchenne Muscular Dystrophy (DMD) using in situ hybridization and immunohistochemistry. They demonstrated that TNF-␣ mRNA and protein were present in muscle fibers in all three disease entities but were rare or absent in neurologic disorders or normal controls. The proportion of TNF-␣– positive fibers correlated with the proportion of regenerating fibers (those that were positive for the developmental form of myosin heavy chain, MHC-d) suggesting that TNF-␣ is produced and expressed by muscle fibers and is associated with regeneration. Although the role of TNF in the pathogenesis of inflammatory myopathies remains uncertain, anecdotal evidence of the efficacy of anti-TNF therapy in inflammatory myopathies has appeared. We believe that such 702 Myositis and myopathies evidence must be heavily discounted because of a very strong reporting bias towards positive outcomes in the early application of almost any new therapy [16]. The apparent development of clinical polymyositis in a patient with long-standing RA who underwent infliximab therapy merits notice. Although the patient was clinically asymptomatic with respect to polymyositis, anti-Jo-1 antibodies were present before the initiation of antiTNF-␣ therapy [17]. Thus anti-TNF-␣ agents should be administered with caution in patients with myositisspecific as well as antinuclear antibodies even in the absence of a clinical myopathy. Co-stimulatory molecules One of the most exciting recent discoveries is the finding of the co-stimulatory molecule B7-H1 expression in cultured human myoblasts in the presence of INF-␥ as well as its presence in inflammatory myopathy muscle specimens. Weindl et al. [18•] examined the pathophysiologic significance of B7-H1 by examining muscle biopsies from patients with IIM as well as those with a noninflammatory myopathy and normal controls for B7-H1 expression by immunohistochemistry. MHC Class I expression and anti-CD28 were assessed as well. Nonmyopathic and noninflammatory myopathy specimens had no evidence of B7-H1 expression, whereas B7-H1 expression was detected in almost all inflammatory myopathy specimens. The B7-H1 positive muscle fibers were in direct contact with T cells, and they were found at the juxtaposition of inflammatory cells and non-necrotic damaged muscle fibers. The authors contend that the role of B7-H1 may be a protective mechanism activated in response to damage of MHC-expressing target cells provoked by INF-␥. Human cells express inducible co-stimulator ligand (ICOSL), another functional co-stimulatory member of the B7 family. ICOSL interacts with its receptor, ICOS, on activated T cells to regulate CD4 and CD8 T-cell responses. Wiendl et al [19•] showed that normal fibers constitutively expressed low levels of ICOSL while muscle fibers in patients with inflammatory myopathies demonstrated markedly increased ICOSL expression. The ICOSL-ICOS interactions may play a part in antigen presentation in the pathogenesis of inflammatory myopathies. Myosin heavy chain The utility of MHC Class I staining in evaluating myopathies is receiving considerable attention. Van der Pas et al. [20] proposed to determine whether MHC Class I antigen expression is upregulated in the sarcolemma in patients with IIM, and if so, whether this upregulation could be used as an additional diagnostic tool. They concluded that detection of sarcolemmal MHC Class I is a valid test for IIM and is not affected by immunosuppressive therapy used for less than 4 weeks. About 4% of biopsies from patients with a dystrophy were positive. In a smaller study, Civatte et al. [21] further investigated the value of MHC Class I detection in muscle biopsies of patients believed to have DM clinically despite having noninformative muscle biopsies. They found abnormal sarcolemmal expression of class I MHC regardless of whether or not the muscle biopsy demonstrated histopathology consistent with DM. In patients with typical biopsy features, class I expression was observed in nearly all muscle fibers but was strongest in perifascicular areas or was restricted to perifascicular atrophic fibers. In all muscle biopsies of DM patients without typical DM features, only some perifascicular fibers expressed class I MHC. They concluded that abnormal perifascicular MHC class I expression may aid in diagnosis in patients with clinical DM in the absence of conclusive histopathologic evidence of the disease. Cytokine/cell adhesion/signaling molecules/chemokines/receptors Both angiogenesis and leukocyte recruitment play a critical role in chronic inflammation, and chemokines have an essential function in these processes. De Paepe et al. [22] hypothesized that the CXCR4/SDF-1 interaction represented a regulatory system involved in the accumulation, migration, and activation of lymphocytes important in the immunopathogenesis of IIM. They demonstrated that CXCR chemokine receptors levels are elevated in IIM, suggesting either that the CXCR4positive cells are selectively recruited to sites of inflammation via local SDF-1 secretion or that membranous CXCR4 secretion is locally upregulated. Thus the SDF1/CXCR4 interaction may be a useful therapeutic target. Autoreactive T cells are important mediators of tissue injury and potential targets for intervention in human autoimmune diseases including polymyositis. In a technically fascinating and impressive paper, Hofbauer et al. [23••] successfully characterized the TCR of autoaggressive T cells over time in biopsies of patients with myositis. This important technique has applicability to other autoimmune inflammatory disorders as well. Lindvall’s group showed that the LFA-1/VLA-4 ratio was lower in patients with PM compared with patients with noninflammatory myopathy or normal controls [24]. They suggest that the VLA-4/VCAM-1 system is an important part of chronic T-cell inflammation of the muscle. S-protein and CD-59 are complement regulators that bind to and inactivate the complement membrane attack complex (MAC). In addition to the deposition of MAC in necrotic fibers, MAC has been found in non-necrotic muscle fibers in inflammatory myopathies and muscular dystrophies where it is believed that MAC deposition may play a role in muscle damage [25]. Louboutin et al. Myositis Christopher-Stine and Plotz 703 [26] characterized the putative role of S-protein and CD59 in DMD and PM—two diseases where MAC deposition has been found. They showed that CD59 disappears from the sarcolemma when fiber necrosis occurs; thus, it cannot inhibit the formation of MAC as it usually does. Secondly, their results suggested that S-protein was not able to prevent the full assembly of MAC in necrotic fibers of DMD and PM patients; rather it could inactivate MAC deposition inside necrotic fibers or clear the MAC-targeted muscle fibers. Oxidative stress Oxidative stress has been implicated in the pathogenesis of inflammatory and noninflammatory muscle diseases. Semicarbazide-sensitive amine oxidase (SSAO) deaminates aromatic and aliphatic primary amines. In the catalytic reaction, oxygen is consumed, and hydrogen peroxide, aldehydes, and ammonia are the final products. SSAO is present in plasma and associated with mammalian tissue membranes, but it is found in very low levels in human skeletal muscle [27•]. Testing the idea that deamination is enhanced in inflammatory myopathies and that SSAO’s contribution results in an additional source of oxidative stress in myopathies, Olive et al. [28] demonstrated the overexpression of SSAO in diseased skeletal muscle. Transglutaminase 2 overexpression Knowing that transglutaminase 2 (TGase 2) is overexpressed in sporadic inclusion body myositis (sIBM), Choi et al. [29] concluded from their prior work with sIBM that an increase in transglutaminase 2 may also be present in other inflammatory myopathies, such as PM and DM. Choi, who had previously shown aberrant expression of TGAse 2 is associated with inclusion body formation in sIBM, considered that overexpression of TGase 2 might be a universal feature of IIM. Immunocytochemistry and quantitative RT-PCR on the muscle tissue of patients with Duchenne muscular dystrophy (DMD) and normal controls as well as patients with PM and DM uncovered an increased level of TGAse 2 expression only in PM and DM tissues. The jump to the conclusion that TGase 2 inhibition might open a therapeutic avenue in the idiopathic inflammatory myopathies seems premature. Autoimmunity: Jo-1 Anti-Jo-1, directed against the ubiquitous intracellular enzyme, histidyl tRNA synthetase, is the most common myositis-specific antibody. Although it is often described in concert with interstitial lung disease, it has rarely been described with concomitant malignancy. A recent report of a case of poorly differentiated adenocarcinoma in a patient who was anti-Jo-1 positive [30] raises new possibilities about the source of the antigens driving myositisspecific autoantibodies, the relation of the myopathy to the associated tumor, and the order of events, since the autoantibodies and the tumor both antedate the onset of clinical myopathy. Genetic susceptibility Specific HLA subtypes are believed to confer increased risk for developing PM and DM. These include HLADRB1*03 in Caucasians and HLA-DRB1*14 in Koreans [31,32]. It is not known, however, whether this association is primary or merely represents an association with other genes in the HLA region. Hassan et al. [33] conducted a study in 65 adult patients with PM or DM to analyze associations between individual genotypes in the HLA region as well as various haplotypes including the following genetic markers: SNPs in HLA-DRB1 and TNF genes as well as microsatellite markers in TNF and MICA genes. Their observations suggested that the ancestral haplotype (A1;B8;DRB1*03) along with the TNF2 allele, as opposed to the HLA-DR3 gene itself, is an important susceptibility factor for the development of PM and DM. Lampe et al. [34] found a significant increase in frequency of the following HLA alleles in patients with sIBM compared with normal controls: A*03, B*08, DRB1*03, and DQB1*05, with DRB1*03 having the most statistically significant increase as compared with normals. The authors contend, as others have before, that HLA typing may help to distinguish between IBM subtypes and perhaps may predict who will benefit from future therapeutic interventions. Because of the similarities observed in the skin and muscle of patients with chronic graft versus host disease (cGVHD) and dermatomyositis, particularly juvenile DM, the pathogenesis is thought to be similar. Microchimerism—the presence of cells acquired by transplacental transfer from fetus to mother or mother to fetus— has been observed in both peripheral blood and muscle cells of patients with IIM. HLA-DQA1*0501 is believed to confer increased risk for these diseases in some populations. Although HLA-DQA1*0501 has been associated with microchimerism [35], Artlett et al. [36] were unable to show that DQA1*0501 was associated with microchimerism in T lymphocytes or whole blood DNA in patients with SSc, juvenile IIM, or healthy controls. Thus, the HLADQA1 alleles do not appear to play a role in the persistence of microchimerism inpatients with autoimmune myopathies. Inclusion body myositis Autoimmunity It is debated whether sIBM is primarily immunemediated; some experts, particularly neurologists, prefer the name inclusion body myopathy. A recent case describing the concomitant findings of anti-PM-SCl antibodies in a patient with IBM supports a role for autoimmunity pathogenesis of IBM [37]. In addition, IBM has 704 Myositis and myopathies been described in concert with autoimmune rheumatic diseases, albeit rarely. Most recently, the combination of SLE, Sjogren syndrome, and IBM was reported for the first time [38]. The patient had SLE for over a decade when she presented with muscle weakness. Electron microscopy demonstrated intranuclear and intracytoplasmic tubulofilamentous inclusions; the patient improved with high-dose methotrexate therapy. Muntzing et al. [39] demonstrated the presence of clonal restriction of TCR expression in muscle-infiltrating lymphocytes in IBM. Identical T-cell clones predominate in different muscles and exist for many years, suggesting an antigen-driven inflammatory reaction in IBM. Protein folding and trafficking The “unfolded protein response” (UPR) is a functional mechanism whereby cells protect themselves from endoplasmic reticulum (ER) stress by assuring proper folding and preventing buildup of unfolded proteins in the ER. This is accomplished with the help of molecular chaperones such as calnexin, calreticulin, GRP94, BiP/GRP78, and ERp72. Expression and immunolocalization of these proteins was studied in patients with sIBM and controls, and in amyloid--precursor protein (AßPP) overexpressing cultured human muscle fibers [40]. All five ER chaperones physically associated with AlPP in sIBM muscle, implying that they may play a role in AßPP folding and processing. The lysosomal system of muscle fibers may also play a critical role in rimmed vacuole formation [41]. Work by Kumamoto et al. [42] showed that the transport of newly synthesized lysosomal enzymes via the Golgi apparatus and autophagic vacuole formation in the lysosome system is activated in sIBM. This was demonstrated by the observation of clathrin and M6PR, which facilitate receptor-mediated intracellular transport inside rimmed vacuoles and in the sarcoplasm of vacuolated or nonvacuolated fibers, but not in control specimens. PrPSc, a hallmark of prion diseases such as spongiform encephalopathies, was demonstrated to be a prominent constituent in sporadic IBM muscle tissue of a patient with concomitant Creutzfeldt-Jakob Disease (CJD)[43]. The existence of muscle disease in subtypes of CJD may deserve further systematic investigation, as distinct glycotyopes of PrSc may be present in muscle and brain. The muscle glycotype in this case report resembled that found in vCJD brain. Although it is attractive to connect vacuolar trafficking abnormalities of glycoproteins to IBM because of the known genetic lesion in one type of hereditary IBM (see below), for the moment, it seems safest to consider these observations as evidence of yet another group of proteins being trapped in IBM inclusions. UDP-N-acetylglucosamine-2-epimerase/N-acetylmannosamine kinase (GNE) mutation The genetic absence of GNE activity leads to muscle weakness in hereditary inclusion body myositis (HIBM), perhaps by interfering with the trafficking of glycoproteins. Huizing et al. [44•] examined the glycosylation status of ␣-dystroglycan (a central protein of skeletal muscle dystrophin) in muscle biopsies of four HIBM patients and found absent or markedly reduced ␣-dystroglycans using antibodies specific for -dystoglycan and laminin ␣-2. They suggest that HIBM may therefore be a “dystroglycanopathy,” similar to the process in congenital muscular dystrophies—perhaps providing one mechanism for the muscle weakness of HIBM patients. In an important series of observations, inflammation has now been clearly recognized in hereditary IBM. Yabe et al. [45••] reported two cases of distal myopathy with rimmed vacuoles (DMRV) in a Japanese family in which inflammation (an unusual feature of DMRV) was present as well as a compound heterozygous mutation in their GNE gene. Mutations associated with DMRV in this study were localized to the kinase domain. An additional novel homozygous mutation was discovered in a nonJewish Iranian population with quadriceps-sparing myopathy consistent with adult onset hIBM: G-to-A mutation in exon 7 changing valine to isoleucine in the epimerase domain of GNE [46]. Muscle inflammation was present in this case as well. These important observations blur the boundary between purely hereditary and sporadic IBM, in particular by raising the possibility that the inflammation seen in apparently sporadic IBM might be a downstream secondary event following damage induced by as-yet unrecognized genetic mutations. The Middle East cluster of hIBM is the result of a founder mutation with incomplete penetrance and is not limited only to the Jewish population. One hundred twenty-nine patients in 55 families with a known history of hIBM were homozygous for the M712T mutation, initially described as the “Persian Jewish Mutation” [47]. Argov et al. [47] have found that the phenotypic spectrum is wider than initially thought. In a study by Del Bo et al., genetic analysis of the GNE gene in an Italian family with autosomal recessive h-IBM demonstrated two novel mutations: a heterozygous deletion involving exons 1–9 and the missense R162C mutation [48]. The quadriceps weakness was apparently distinct from that found in the quadriceps sparing nonPersian Jewish families with a different GNE mutation. Hereditary IBM with early-onset Paget disease of bone and frontotemporal dementia (IBMPFD) is a rare form of hereditary myopathy. Its clinical attributes distinguish it from the other forms of hIBM linked to chromosome 9. Myositis Christopher-Stine and Plotz 705 Watts et al. [49] found no relation of GNE mutations with IBMPFD, confirming genetic heterogeneity with IBM2. lungs and the skeletal muscle to account for the high prevalence of concomitant pathology and the autoantibodies—directed to aminoacyl-tRNA synthetases—found in patients with both lung and muscle involvement. 9 Cottin V, Thivolet-Bejui F, Reynaud-Gaubert M, et al.: Interstitial lung disease in amyopathic dermatomyositis, dermatomyositis and polymyositis. Eur Respir J 2003, 22:245–250. 10 High WA, Cohen JB, Murphy BA, Costner MI: Fatal interstitial pulmonary fibrosis in anti-Jo-1-negative amyopathic dermatomyositis. J Am Acad Dermatol 2003, 49:295–298. 11 Bronner IM, Hoogendijk JE, Wintzen AR, et al.: Necrotising myopathy, an unusual presentation of a steroid-responsive myopathy. J Neurol 2003, 250:480–485. Other genetic evidence The muscle morphology in X-EDMD is not pathognomonic; rather, there are nonspecific myopathic changes including endomysial connective tissue proliferation, fiber splitting, type II fiber predominance, and type I fiber atrophy. A recent paper reported the co-existence of x-linked Emery-Dreifuss muscular dystrophy (EDMD) and IBM [50]. Thus typical IBM morphologic features may be found in other neuromuscular disorders. Conclusion Although the pathogenesis of the inflammatory myopathies remains obscure, great strides over the past year have placed us closer to understanding the etiologies of these diverse disease entities. We have deepened our understanding that although these diseases may share some components of the clinical phenotype as well as some serologic similarities, they differ on a molecular and cellular level. The inflammatory myopathies result from a combination of interactions between environmental and genetic risk factors, and the need for definitive, safer therapeutic options in inflammatory myopathies makes the search for defining detailed pathogenesis of inflammation and muscle fiber damage at the molecular level essential. De Bleecker J, Vervaet V, Van den Bergh P: Necrotizing myopathy with microvascular deposition of the complement membrane attack complex. Clin Neuropathol 2004, 23:76–79. The papers of Bronner [11] and De Bleecker resemble earlier observations on the necrotizing myopathy with little or no inflammation seen in patients with anti-SRP autoantibodies. 12 • Okada S, Weatherhead E, Targoff IN, et al.: Global surface ultraviolet radiation intensity may modulate the clinical and immunologic expression of autoimmune muscle disease. Arthritis Rheum 2003, 48:2285–2293. A group of clinicians from around the around the world with expertise in myositis joined together by Dr. Fred Miller and his colleagues in the National Institute of Environmental Health Sciences at NIH have made an excellent advance in providing a plausible model for how an environmental factor might influence the phenotype of a disease with a strong genetic component. Their success depended upon starting with a good hypothesis. 13 •• 14 Werth VP, Bashir M, Zhang W: Photosensitivity in rheumatic diseases. J Investig Dermatol Symp Proc 2004, 9:57–63. 15 Kuru S, Inukai A, Kato T, et al.: Expression of tumor necrosis factor-alpha in regenerating muscle fibers in inflammatory and non-inflammatory myopathies. Acta Neuropathol (Berl) 2003, 105:217–224. 16 Hengstman GJ, van den Hoogen FH, Barrera P, et al.: Successful treatment of dermatomyositis and polymyositis with anti-tumor-necrosis-factor-alpha: preliminary observations. Eur Neurol 2003, 50:10–15. 17 Musial J, Undas A, Celinska-Lowenhoff M: Polymyositis associated with infliximab treatment for rheumatoid arthritis. Rheumatology (Oxford) 2003, 42:1566–1568. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest Andersson J, Englund P, Sunnemark D, et al.: CBA/J mice infected with Trypanosoma cruzi: an experimental model for inflammatory myopathies. Muscle Nerve 2003, 27:442–448. Despite the apparent remoteness of this parasite-induced disease from the apparently autoimmune pathology of human idiopathic inflammatory myopathy, this model mimics the pathologic picture closer than most others. It may, however, prove too remote and unfamiliar to be attractive to other investigators. 1 •• Wiendl H, Mitsdoerffer M, Schneider D, et al.: Human muscle cells express a B7-related molecule, B7-H1, with strong negative immune regulatory potential: a novel mechanism of counterbalancing the immune attack in idiopathic inflammatory myopathies. FASEB J 2003, 17:1892–1894. This is a follow-up of important earlier observations on this newly recognized costimulatory path. 18 • Wiendl H, Mitsdoerffer M, Schneider D, et al.: Muscle fibres and cultured muscle cells express the B7.1/2-related inducible co-stimulatory molecule, ICOSL: implications for the pathogenesis of inflammatory myopathies. Brain 2003, 126:1026–1035. Another possibly important co-stimulatory pair identified by this group adds expected complexity to the picture of pathogenesis. 19 • 20 van der Pas J, Hengstman GJ, ter Laak HJ, et al.: Diagnostic value of MHC class I staining in idiopathic inflammatory myopathies. J Neurol Neurosurg Psychiatry 2004, 75:136–139. Petersen LR, Marfin AA: West Nile virus: a primer for the clinician. Ann Intern Med 2002, 137:173–179. 21 4 Douche-Aourik F, Berlier W, Feasson L, et al.: Detection of enterovirus in human skeletal muscle from patients with chronic inflammatory muscle disease or fibromyalgia and healthy subjects. J Med Virol 2003, 71:540–547. Civatte M, Schleinitz N, Krammer P, et al.: Class I MHC detection as a diagnostic tool in noninformative muscle biopsies of patients suffering from dermatomyositis (DM). Neuropathol Appl Neurobiol 2003, 29:546–552. 22 5 Leff RL, Love LA, Miller FW, et al.: Viruses in idiopathic inflammatory myopathies: absence of candidate viral genomes in muscle. Lancet 1992, 339:1192–1195. De Paepe B, Schroder JM, Martin JJ, et al.: Localization of the alphachemokine SDF-1 and its receptor CXCR4 in idiopathic inflammatory myopathies. Neuromuscul Disord 2004, 14:265–273. 2 Smith RD, Konoplev S, DeCourten-Myers G, Brown T: West Nile virus encephalitis with myositis and orchitis. Hum Pathol 2004, 35:254–258. 3 6 Chevrel G, Borsotti JP, Miossec P: Lack of evidence for a direct involvement of muscle infection by parvovirus B19 in the pathogenesis of inflammatory myopathies: a follow-up study. Rheumatology (Oxford) 2003, 42:349–352. Fathi M, Dastmalchi M, Rasmussen E, et al.: Interstitial lung disease, a common manifestation of newly diagnosed polymyositis and dermatomyositis. Ann Rheum Dis 2004, 63:297–301. A fine report of a large experience with a remarkably high proportion of patients with lung involvement. 7 • Schnabel A, Reuter M, Biederer J, et al.: Interstitial lung disease in polymyositis and dermatomyositis: clinical course and response to treatment. Semin Arthritis Rheum 2003, 32:273–284. Another nicely studied series with a relatively high incidence of lung involvement. It seems likely that there will turn out to be common microenvironmental factors in the 8 • Hofbauer M, Wiesener S, Babbe H, et al.: Clonal tracking of autoaggressive T cells in polymyositis by combining laser microdissection, single-cell PCR, and CDR3-spectratype analysis. Proc Natl Acad Sci USA 2003, 100:4090– 4095. An impressive array of highly sensitive techniques have been brought together to explore important questions about individual immune cells that inhabit the inflammatory lesions in myositis. A tour de force. 23 •• 24 Lindvall B, Dahlbom K, Henriksson KG, et al.: The expression of adhesion molecules in muscle biopsies: the LFA-1/VLA-4 ratio in polymyositis. Acta Neurol Scand 2003, 107:134–141. 25 Sewry CA, Dubowitz V, Abraha A, et al.: Immunocytochemical localisation of complement components C8 and C9 in human diseased muscle. The role of complement in muscle fibre damage. J Neurol Sci 1987, 81:141–153. 26 Louboutin JP, Navenot JM, Rouger K, Blanchard D: S-protein is expressed in 706 Myositis and myopathies necrotic fibers in Duchenne muscular dystrophy and polymyositis. Muscle Nerve 2003, 27:575–581. Andres N, Lizcano JM, Rodriguez MJ, et al.: Tissue activity and cellular localization of human semicarbazide-sensitive amine oxidase. J Histochem Cytochem 2001, 49:209–217. This is an intriguing new window on the very local events where the damage is occurring. 27 • 28 Olive M, Unzeta M, Moreno D, Ferrer I: Overexpression of semicarbazidesensitive amine oxidase in human myopathies. Muscle Nerve 2004, 29:261– 266. 29 Choi YC, Kim TS, Kim SY: Increase in transglutaminase 2 in idiopathic inflammatory myopathies. Eur Neurol 2004, 51:10–14. 30 Watkins J, Farzaneh-Far R, Tahir H, et al.: Jo-1 syndrome with associated poorly differentiated adenocarcinoma. Rheumatology (Oxford) 2004, 43:389–390. 31 Hausmanowa-Petrusewicz I, Kowalska-Oledzka E, Miller FW, et al.: Clinical, serologic, and immunogenetic features in Polish patients with idiopathic inflammatory myopathies. Arthritis Rheum 1997, 40:1257–1266. 32 Rider LG, Shamim E, Okada S, et al.: Genetic risk and protective factors for idiopathic inflammatory myopathy in Koreans and American whites: a tale of two loci. Arthritis Rheum 1999, 42:1285–1290. 33 Hassan AB, Nikitina-Zake L, Sanjeevi CB, et al.: Association of the proinflammatory haplotype (MICA5.1/TNF2/TNFa2/DRB1*03) with polymyositis and dermatomyositis. Arthritis Rheum 2004, 50:1013–1015. 34 Lampe JB, Gossrau G, Kempe A, et al.: Analysis of HLA class I and II alleles in sporadic inclusion-body myositis. J Neurol 2003, 250:1313–1317. 35 clonal expansions of muscle-infiltrating T cells persist over time. Scand J Immunol 2003, 58:195–200. 40 Vattemi G, Engel WK, McFerrin J, Askanas V: Endoplasmic reticulum stress and unfolded protein response in inclusion body myositis muscle. Am J Pathol 2004, 164:1–7. 41 Tsuruta Y, Furuta A, Furuta K, et al.: Expression of the lysosome-associated membrane proteins in myopathies with rimmed vacuoles. Acta Neuropathol (Berl) 2001, 101:579–584. 42 Kumamoto T, Ueyama H, Tsumura H, et al.: Expression of lysosome-related proteins and genes in the skeletal muscles of inclusion body myositis. Acta Neuropathol (Berl) 2004, 107:59–65. 43 Kovacs GG, Lindeck-Pozza E, Chimelli L, et al.: Creutzfeldt-Jakob disease and inclusion body myositis: abundant disease-associated prion protein in muscle. Ann Neurol 2004, 55:121–125. Huizing M, Rakocevic G, Sparks SE, et al.: Hypoglycosylation of alphadystroglycan in patients with hereditary IBM due to GNE mutations. Mol Genet Metab 2004, 81:196–202. This study is an imaginative approach to make the connection between a known genetic lesion and a distant clinical symptom. 44 • 45 Yabe I, Higashi T, Kikuchi S, et al.: GNE mutations causing distal myopathy with rimmed vacuoles with inflammation. Neurology 2003, 61:384–386. •• An interesting and welcome complexity that suggests that it is highly likely that inflammation in many circumstances in muscle disease will one day be discovered to be a secondary event. 46 Krause S, Schlotter-Weigel B, Walter MC, et al.: A novel homozygous missense mutation in the GNE gene of a patient with quadriceps-sparing hereditary inclusion body myopathy associated with muscle inflammation. Neuromuscul Disord 2003, 13:830–834. Lambert NC, Evans PC, Hashizumi TL, et al.: Cutting edge: persistent fetal microchimerism in T lymphocytes is associated with HLA-DQA1*0501: implications in autoimmunity. J Immunol 2000, 164:5545–5548. 47 Argov Z, Eisenberg I, Grabov-Nardini G, et al.: Hereditary inclusion body myopathy: the Middle Eastern genetic cluster. Neurology 2003, 60:1519– 1523. 36 Artlett CM, O’Hanlon TP, Lopez AM, et al.: HLA-DQA1 is not an apparent risk factor for microchimerism in patients with various autoimmune diseases and in healthy individuals. Arthritis Rheum 2003, 48:2567–2572. 48 Del Bo R, Baron P, Prelle A, et al.: Novel missense mutation and large deletion of GNE gene in autosomal-recessive inclusion-body myopathy. Muscle Nerve 2003, 28:113–117. 37 Selva-O’Callaghan A, Mijares-Boeckh-Behrens T, Labrador-Horrillos M, et al.: Anti-PM-Scl antibodies in a patient with inclusion body myositis. Rheumatology (Oxford) 2003, 42:1016–1018. 49 Watts GD, Thorne M, Kovach MJ, et al.: Clinical and genetic heterogeneity in chromosome 9p associated hereditary inclusion body myopathy: exclusion of GNE and three other candidate genes. Neuromuscul Disord 2003, 13:559– 567. 38 Derk CT, Vivino FB, Kenyon L, Mandel S: Inclusion body myositis in connective tissue disorders: case report and review of the literature. Clin Rheumatol 2003, 22:324–328. 50 39 Muntzing K, Lindberg C, Moslemi AR, Oldfors A: Inclusion body myositis: Fidzianska A, Rowinska-Marcinska K, Hausmanowa-Petrusewicz I: Coexistence of X-linked recessive Emery-Dreifuss muscular dystrophy with inclusion body myositis-like morphology. Acta Neuropathol (Berl) 2004, 107:197– 203. Have recent immunogenetic investigations increased our understanding of disease mechanisms in the idiopathic inflammatory myopathies? Hector Chinoy, William E.R. Ollier and Robert G. Cooper Purpose of review The idiopathic inflammatory myopathies (IIM) continue to provide a challenge given the variable effectiveness of the available treatments, and immunogenetic studies are ongoing to further elucidate IIM disease mechanisms. This review examines how recent research has improved our understanding of the mechanisms that lead to IIM. Recent findings HLA-DRB1 studies in a large homogenous cohort of UK Caucasian patients have confirmed that polymyositis (PM) and dermatomyositis (DM) are not genetically identical diseases while other studies have shown that tumor necrosis factor alpha is genetically implicated in disease susceptibility. Some remarkable results from an international collaboration, correlating gene-environment interactions, clearly suggest that ultraviolet light is capable of modulating both clinical and immunologic features of IIMs. Studies on microchimerism are unraveling interesting associations in juvenile DM patients, and bolstering the hypothesis that myositis may be an ‘allo-immune’ disease. mRNA gene expression profiling is helping to increase our understanding of myositis pathogenesis, whilst animal models have provided new information on the roles of Th1 responses and nitric oxide synthase in muscle disease. New candidate genes have been examined in inclusion body myositis (IBM), and a novel gene transfer experiment has been conducted, which led to significant changes in expression of the IBM phenotype. Summary Improving the understanding of the immunogenetics and immunopathogenesis of the IIMs may in the future provide novel therapeutic targets, and thus improve outcomes in these difficult diseases. Keywords idiopathic inflammatory myopathies, immunogenetics, polymyositis, dermatomyositis, inclusion body myositis, HLA, haplotypes, microchimerism Curr Opin Rheumatol 16:707–713. © 2004 Lippincott Williams & Wilkins. Rheumatic Diseases Centre, Hope Hospital, Salford, UK Correspondence to Robert G. Cooper, Rheumatic Diseases Centre, Hope Hospital, Salford M6 8HD, UK Tel: +44 (0)161 206 4367; e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:707–713 © 2004 Lippincott Williams & Wilkins 1040–8711 Introduction The idiopathic inflammatory myopathies (IIMs) are a heterogeneous group of potentially serious rare diseases, defined by the presence of acquired muscle inflammation and weakness. Polymyositis (PM) and dermatomyositis (DM) are the subtypes most often seen by rheumatologists. Although corticosteroids, immunosuppressive agents, and intravenous immunoglobulins can all be effective in managing PM/DM, the response to these therapies is variable, and often disappointing. Patients thus occasionally die from their disease, while survivors frequently remain disabled through persisting weakness or lung fibrosis. Given the limited effectiveness of available agents, new and more potent therapies are clearly needed. However, to facilitate the development of novel therapies, the etiopathogenic mechanisms underlying these conditions require further elucidation. The IIMs are rare diseases, with an annual incidence ranging 2.18 to 7.7 cases per million [1]; so research into these mechanisms has proved difficult. It is becoming increasingly obvious that genetic factors are involved [2]. The previous evidence for a genetic basis in IIMs, as extensively reviewed in 2000 by Shamim et al. [3], has largely come from candidate gene studies, as the rarity of IIMs has precluded the use of more robust genetic methods, such as twin studies, whole genome scans, and transmission disequilibrium testing [4]. However, multiple case reports where two or more members are affected in one family [3], clearly suggest a familial predisposition for developing IIMs. Indeed, Rider et al. [5] confirmed that HLA-DRB1*0301 and homozygosity of HLA-DQA1 represent risk factors for developing familial IIMs. Candidate gene studies in nonfamilial IIM have mainly concentrated on the HLA class II region, confirming that HLA-DRB1*0301 and the linked allele HLA-DQA1*0501 are risk factors for developing IIMs in Caucasians, but not in Mesoamerican Mestizos, Koreans, or Japanese populations [6–10]. Gene polymorphisms other than HLA class II are also associ707 708 Myositis and myopathies ated with IIMs, including those of the IL-1 receptor antagonist [11], tumor necrosis factor alpha (TNF␣) [12], and Gm/Km allotypes [8]. HLA-related differences in polymyositis/dermatomyositis Most previous IIM candidate gene studies have analyzed adult and juvenile PM/DM patient results together, and some studies even included results from patients with inclusion body myositis (IBM) [3]. Such ‘pooling’ reflects the sample size problems outlined, and was presumably undertaken to gain power during statistical comparisons of IIM patients with control subjects. Given the phenotypic differences (ie, of clinical, serological, and pathologic features) between IIM subtypes, a more logical approach would be to compare and contrast, rather than group, these diseases. To get over the sample size problems, and thus to address the question of whether PM and DM are genetically the same with respect to HLA class II, we recently coordinated a nationwide collaborative study of 59 physicians, constituting the UK ‘Adult Onset Myositis Immunogenetic Collaboration’ (AOMIC), which recruited 110 PM and 98 DM UK Caucasian patients over 4 years (Chinoy et al, unpublished data). These patients’ HLA-DRB1 data were compared with those of 537 ethnically matched controls. The results confirmed that HLA-DRB1*03 was a risk factor for PM (odds ratio [OR] 4.0, 95% confidence interval [CI] 2.6–6.1) (Table 1). However, there was also a significant protective effect of HLA-DRB1*07 in PM versus controls (OR 0.3, 95% CI 0.4–0.6). The results for DM patients were different, as the association with HLADRB1*03 was considerably weaker than for PM (OR 2.0, 95% CI 1.3–3.1) and, by contrast with PM, DRB1*07 represented a risk factor versus controls (OR 1.8, 95% CI 1.2–2.9). These results suggest that, at least in a UK Caucasian population, HLA-DRB1 not only plays a role in governing PM/DM disease susceptibility, through association with DRB1*03, but may also govern myositis disease phenotype, through association with DRB1*07. These alleles may be a marker for another contributory factor (eg, a different allele in linkage disequilibrium forming part of a broader haplotype, or an autoantibody). Autoantibodies and ‘elemental disorders’ It was previously thought that genetically predisposed individuals may only develop their autoimmune disease after certain interactions with environmental triggers [13,14], so is there any recent evidence to support this? Two previous studies suggested that decreasing latitude exerts an influence on the proportion of a myositis population with DM rather than PM [8,15]. The findings of a more recent paper by Okada et al. [16••] are thus of great interest. In studying 919 IIM patients in 15 global locations for 13 different climatic variables, surface ultraviolet (UV) light intensity proved the strongest contributor to the relative proportion of DM compared with PM (r = 0.939, P < 4 × 107), and to the relative proportion of anti-Mi-2 antibodies (r = 0.69, P = 0.02) (Fig. 1). It thus appears that the greater an individual’s exposure to sunlight, the greater the likelihood that, if that individual did develop myositis, it would be of anti-Mi-2 positive DM in type. These remarkable results suggest that an environmental factor may be capable of modulating the expression of both the clinical and immunologic features of the resulting myositis. The authors speculated that sunlight achieves this by playing an etiological role in the development of Mi-2 antibodies, and thus DM, via mechanisms of UV-induced immune dysregulation [16••]. The presence of anti-Mi-2 antibodies in DM patients are very strongly associated with HLA-DRB1*0701, with Mierau et al. [17] demonstrating an OR of 22 versus controls, in a Caucasian population, and Shamim et al. [8] demonstrating an association in Caucasians and Mexican Mestizos. Miller describes the concept of ‘elemental disorders’, where autoimmune diseases as we understand them, are the result of a specific pathogenesis due to interactions between genetic and elemental environmental risk factors to produce unique sign-symptom complexes [18]. Nonclassical HLA and cytokine genes TNF␣ is a pro-inflammatory cytokine that plays a major role in immune response regulation, and is encoded within the MHC class III region. The TNF␣ promoter single nucleotide polymorphism (SNP) at position −308, resulting from a G to an A substitution, is termed TNF2 (−308A), and associations with this allele are found in juvenile DM (JDM) [12] and adult DM [19]. A recent JDM gene expression profile study led to the proposal of a novel JDM pathogenesis model, which encompasses antiviral, ischemic, and degeneration/regeneration cascades, in which TNF␣ is thought to be a key molecule [20]. Certainly, in JDM, TNF2 (−308A) is associated with disease chronicity, pathologic calcification [19], and increased circulating concentrations of an anti- Table 1. Immunogenetic risk factors in UK AOMIC cohort Controls (n = 537) HLA DRB1*03 DRB1*07 n (%) 151 (28.1) 129 (24.0) Polymyositis (n = 110) n (%) 67 (60.9) 9 (8.2) p Dermatomyositis (n = 98) pcorr −11 3 × 10 1 × 10−04 −10 4 × 10 0.001 OR (95%CI) n (%) p pcorr OR (95%CI) 4.0 (2.6–6.1) 0.3 (0.4–0.6) 43 (43.9) 36 (36.7) 0.002 0.008 0.022 0.100 2.0 (1.3–3.1) 1.8 (1.2–2.9) OR, odds ratio; CI, confidence interval; p, probability; pcorr, corrected for multiple comparisons using Bonferroni adjustment. Disease mechanisms in IIM Chinoy et al. 709 Figure 1. Depiction of correlations in normal muscle, but is found in muscle fibers, inflammatory cells, and capillaries in PM, DM, and IBM [24]. Through alternative splicing of transcripted HLA-G mRNA, at least seven different isoforms have been identified, labeled HLA-G1 to -G7. HLA-G binds CD8+ cells and antigenic peptides, is thought to play an important role in immunotolerance, and more specifically, may help prevent maternal lymphocytes from attacking fetal tissue. Using in vitro models of human myoblasts and TE671 muscle rhabdomyosarcoma cells, two isoforms, transmembranous HLA-G1 and soluble HLA-G5, have been investigated recently [25•]. The HLA-G isoforms inhibited the primary alloreactive response to muscle cells, and inhibited alloreactive lysis mediated by CD4+, CD8+, and natural killer cells. Priming of cytotoxic T cells was also inhibited, and muscle cells were protected from antigen-specific cytotoxic T cell lysis. HLA-G1 and -G5 are therefore powerful inhibitors of the primary and secondary immune responses, and in the future these results could form the basis for developing a novel muscle cell protective agent for reducing IIM cellmediated injury. Microchimerism Correlations between the weighted surface ultraviolet (UV) intensity at each of the global locations and the proportion of DM/DM & PM, and the proportion of anti-Mi-2 autoantibodies/all myositis-specific autoantibodies (MSAs). Reprinted with permission [16••]. Arthritis & Rheumatism © 2003. Reprinted by permission of Wiley-Liss, Inc., a subsidiary of John Wiley & Sons, Inc. angiogenic factor, thrombospondin-1 [21]. TNF2 (−308A) is thought to be part of the common extended haplotype A1;B8;DR3;DQ2, which has been associated with autoimmunity [12]. A recent study compared 65 PM/DM patients to a matched control population, examining the associations of HLA-DRB1*03 and TNF2 (−308A) with two microsatellite markers, MICA5.1 and TNFa2 [22]. The TNF2 (−308A) allele (OR 2.75, 95% CI 1.35–5.61), and the haplotype MICA5.1/TNF2/ TNFa2/DRB1*03 (OR 3.48, 95% CI 1.71–7.09) were both associated with PM/DM compared with controls. In line with other autoimmune diseases, the ancestral haplotype together with the TNF2 (−308A) allele, rather than HLA-DRB1*03 alone, may be an important susceptibility factor for the development of PM/DM [22•]. Muscle fibers in biopsies from IIM patients in vivo, and in cultured muscle cells, express intracellular (cytoplasmic) HLA-G, a nonclassical MHC class I molecule, which exhibits a highly restricted tissue distribution under physiologic conditions [23]. HLA-G is undetectable Microchimerism refers to the low-level persistence of nonself cells due to bidirectional trafficking of fetal/maternal cells during pregnancy [26]. Maternal microchimeric cells (ie, nonhost cells) have been found in the peripheral blood and muscle lesions of juvenile IIM patients (Table 2) [27–33••]. In a recent study of HLA and maternal microchimerism in JDM [33••], maternal chimeric cells were identified in 60 of 72 (83%) JDM patients, versus 5 of 29 (17%) healthy male controls (OR 24, 95% CI 6.9–93.0). All healthy siblings with microchimerism were either HLA-DQA1*0501 positive, or had noninherited (maternal) DQA1*0501 cells present. Maternally derived chimeric cells from JDM patients produced high levels of interferon-gamma (IFN-␥) secreting T cells when exposed to JDM lymphocytes, suggesting an antihost immune response. A further study examined microchimerism in 87 systemic sclerosis (SSc) and 28 JDM patients [32], and demonstrated an increased frequency of maternal microchimerism in SSc (33/47 [70.2%] patients, OR 2.5, 95% CI 0.9–7.6) and JDM (20/28 [71.4%] patients, OR 33.7, 95% CI 5.7–329.8) compared with controls. An increase in HLA-DQA1*0501 was observed in JDM patients positive for maternal microchimerism, although this effect did not reach statistical significance. HLA-DQA1*0501 is also strongly associated with fetal microchimerism in maternal T cells [34], although this has not been demonstrated to date in juvenile IIM. The risk for JDM and other autoimmune diseases may be determined by the HLA genotype of the mother, providing a ‘second hit’ to trigger disease in genetically susceptible individuals [33••]. The inherited HLA genotype may therefore contribute to loss of toler- 710 Myositis and myopathies Table 2. Associations of maternal microchimerism with juvenile IIM patients Sample Method PBMC PBMC PBMC Muscle PBMC† Muscle PBMC NTMA NTMA FISH FISH FISH FISH NTMA PBMC FISH Muscle PBMC PBMC FISH NTMA/Cw NTMA Frequencies of disease vs comparison group Comparison group Associations 3/3 vs 2/7 13/15 vs 5/35 11/15 vs 5/17 12/15 vs 2/10 8/9 vs 0/9 10/10 vs 2/10 25/30 vs 7/39 7/100 22/30 vs 12/39 8/40 16/20 vs 1/10 20/28 vs 2/29 60/72 vs 11/48 5/29 Sibling Sibling Control Control Control Control Sibling Control Sibling Control Control Control Sibling Control p = 0.2* OR 39.0 (5.6–406.8) OR 6.6 (1.1–41.6) OR 16.0 (1.7–202.9) p = 0.0004* p = 0.0007* OR 22.9 (5.7–99.6) OR 66.4 (17.1–278.1) OR 6.1 (1.9–20.6) OR 11.0 (3.2–39.4) OR 36.0 (2.9–1671.8) OR 33.8 (5.7–329.8) OR 16.8 (6.2–46.7) OR 24.0 (6.9–93.0) References [27] [28] [28] [28] [29] [29] [30] [30] [30] [30] [30] [31,32] [33••] [33••] Where odds ratios were not calculated in respective papers cited, the values shown in the table are those derived from data presented in each paper. *Odds ratios not possible as cells have zero values; †CD4+ and CD8+ cells used. OR, odds ratio; PBMC, peripheral blood mononuclear cell; NTMA, nontransmitted maternal HLA-DQA1 allele; FISH, fluorescent in situ hybridization; NTMA/Cw, non-transmitted maternal Cw allele. ance and activation of chimeric cells, thereby initiating the disease process and inflammation. The finding of loss of tolerance is in contrast to other situations where chimerism may be beneficial, such as pregnancy or organ transplantation. An interesting analogy of microchimerism is that of chronic graft-versushost disease (GVHD), where donor cells infiltrate the skin and mucous membranes [35]. Recently, a large cohort of patients undergoing hematopoietic stem cell transplantation over a 30-year period were retrospectively reviewed [36••]. Of the 1859 patients who developed GVHD, 12 also developed a myositis syndrome (GVHD-PM), resembling idiopathic PM in many clinical and laboratory aspects. Not one of the stem cell transplant patients without GVHD developed myositis. As the host’s immune system had been eliminated, the lymphocytic infiltration in the muscle of GVHD-PM patients was assumed to consist of donor cells [35]. The cases of GVHD-PM, and their resemblance to IIM, suggest a common underlying pathogenesis in both conditions, with microchimeric cells in IIM forming the role of donor cells, and bolsters the hypothesis that myositis represents an allo-immune mediated disease [37]. Inclusion body myositis As IBM usually presents with distal, rather than proximal, muscle weakness, and thus mimicking peripheral neuropathy, it is rarely seen by rheumatologists. However, due to presence of inflammation in muscle biopsies, sporadic IBM is considered one of the IIMs and is therefore included in this update. Several HLA and nonHLA loci are already known risk factors for sporadic IBM [3]. HLA class I and II associations have recently been examined in 47 sporadic IBM patients and 29,670 controls [38]. HLA-A*03 (OR 2.6, 95% CI 1.6–4.1), B*08 (OR 2.8, 95% CI 1.6–4.6), DRB1*03 (OR 3.5, 95% CI 2.1–5.6), and DQB1*05 (2.0, 95% CI 1.2–3.3) alleles were all significantly increased compared with controls, even after adjustment for multiple comparisons. The ethnicity of the cases in this study was not stated and these results should thus be interpreted with caution. There are currently no clinical or biochemical parameters that predict the outcome of IBM or response to treatment, and HLA typing may help subgroup IBM patients and predict such parameters. Sporadic IBM muscle biopsies possess structural abnormalities similar to those in brain tissue plaques from Alzheimer’s disease patients, including amyloid- precursor protein (APP), amyloid- (A), and apolipoprotein E (apoE) [39]. Using an adenovirus vector, A gene transfer into normal cultured human muscle fibers induced phenotypic abnormalities similar to those found in IBM, suggesting the likelihood of a key role of APP/A in IBM pathogenesis [40]. The same gene was transferred into muscle fibers from a patient with known sporadic IBM and associated cardiac amyloidosis in whom a Val122Ile transthyretin (TTR) mutation was also expressed [41•]. The resulting overexpression of the A gene amplified the abnormalities found in this patient’s cultured muscle fibers, including accelerated degeneration, inclusions, and vacuole formation, which were over and above those seen in the normal muscle. The TTR mutation could either be a genetic risk factor, or perpetuate the existing IBM. Polymorphisms of two further intracellular amyloid deposits, ApoE and ␣(1)-antichymotrypsin, have been tested in the peripheral blood of 35 sporadic IBM patients [42], but no significant associations or correlations with age of onset were found. An important gene expression profiling study has also found increased expression of amyloid- and ApoE in IBM, but significantly elevated levels of the same genes were also demonstrated in PM and DM, suggesting that accumu- Disease mechanisms in IIM Chinoy et al. 711 lation of such proteins in IBM may be due to posttranscriptional events [43]. T cell receptors Immunohistochemistry studies of biopsies from patients with PM have demonstrated a predominance of CD8+ T lymphocytes invading non-necrotic muscle fibers that express HLA class I on their cell surface [44]. Previous studies have demonstrated clonally expanded T cell receptor (TCR) families in muscle fibers of patients with PM. To characterize the role of these T cells further, a process of CDR3 spectratyping was combined with laser microdissection and single-cell polymerase chain reaction (PCR), to select individual pathogenically relevant T cells from PM muscle biopsies [45]. After examining repeat biopsies in one patient, it was shown that T cell clones could persist for many years. In another patient, several T cells had minor nucleotide changes in the CDR3 region of the T cell receptor, which did not alter the amino acid sequences, thus suggesting the presence of different T cell clones driving a common antigendriven response. Oligoclonal CDR3 spectratype peaks disappeared during immunosuppressive treatment. Recent TCR expression work has also been performed in IBM, again suggesting only a limited number of antigens drive the inflammatory reaction [46]. These pathogenically relevant T cells may represent future epitopes for targeted immunotherapy. Animal models Myositis is thought to be predominantly Th1 driven, initiated by an (unknown) antigen and MHC interaction, leading to T cell expansion, maturation, and subsequent cytokine proliferation (eg, IFN-␥, IL-1, IL-18). Theoretically, dampening down of the Th1 cell response, and switching to a Th2-driven system, producing alternative cytokines (eg, IL-4, IL-6, IL-10), could dampen down the pathogenicity of autoreactive T cells [47]. A strain of nonobese diabetic (NOD) mice has been developed, rendered Th1-deficient by a CD2 promoter driven IFN-␥ receptor -chain transgene [47]. An unexpected consequence was the development of an early lethal myopathy due to a CD8+ T cell-dependent myositis syndrome, characterized by a massive leukocyte inflammatory response in the muscle fibers. By interacting with other genetic components in the NOD model, the inhibition of Th1 responses may conversely have exacerbated certain autoimmune responses. Another way of influencing the inflammatory response in muscle is through nitric oxide (NO), which can be pro- or anti-inflammatory depending on its concentration and locality. A further transgenic mouse model has been developed with muscle-specific overexpression of neuronal nitric oxide synthase (nNOS), driven by a human skeletal muscle actin promoter [48]. Using a rodent hindlimb unloading/reloading model, overexpression of nNOS in- hibited neutrophil production, and prevented any increases in muscle membrane damage. Muscle-derived NO evidently functions as an anti-inflammatory molecule, and may provide a future potential therapeutic target. New and future approaches The work done by Okada et al. [16••] and ourselves demonstrates the advantages, and indeed necessity, of undertaking national/international genetic collaborations. The results of these larger studies illustrate the importance during genetic testing of treating IIM subtypes as discrete, rather than grouped, diagnoses. Due to the rarity of IIMs, only further collaboration is likely to elucidate the complex interactions between genetic and environmental factors. Recently doubt has been cast on the diagnostic category PM, as some patients have been shown to have IBM or muscular dystrophy [49]. mRNA microarray gene expression profiling studies may assist in a more robust molecular reclassification of the IIMs [43], whilst also providing novel molecular insights into the role of genes in IIM subtypes (reviewed in [50•]). It is relatively easy to obtain muscle biopsies, allowing ready analysis of target tissue in IIM patients. Measurements of mRNA levels allow the demonstration of which genes are upregulated or downregulated (albeit without establishing cause and effect), and clarification of which genes to analyze in further candidate gene studies. A recent landmark study examined the molecular profiles of patients with IIMs, showing distinct changes from normal muscle and differing between the IIMs [43]. An important distinguishing feature of DM, compared with PM and IBM, was the increased expression of interferoninducible genes, and this finding was reproduced in a JDM study [20], raising the hypothesis that DM has an antigen-driven pathogenesis, and again supporting the idea that PM and DM are genetically different. In the future, improved techniques and reduced costs may allow the use of powerful high-density SNP arrays to conduct whole genome scans by association [50•–52]. Research continues worldwide into our understanding of the underlying pathogenesis of the IIMs. A basic science approach led to the elucidation of the key role of TNF␣ in RA, and the subsequent use of anti-TNF␣ therapy. By analogy, ongoing collaborative genetic work may help identify key hierarchical molecules implicated in IIM pathology. Acknowledgments The authors thank the Arthritis Research Campaign and the Myositis Support Group (UK), which provided the funds necessary to undertake the AOMIC genetic analysis. We also wish to thank the 59 physicians who contributed to AOMIC. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest 1 Mastaglia FL, Phillips BA: Idiopathic inflammatory myopathies: epidemiology, 712 Myositis and myopathies classification, and diagnostic criteria. Rheum Dis Clin North Am 2002, 28:723–741. tial role of HLA-G on muscle cells and in inflammatory myopathies. Hum Immunol 2003, 64:1050–1056. 2 Plotz PH, Rider LG, Targoff IN, et al.: NIH conference. Myositis: immunologic contributions to understanding cause, pathogenesis, and therapy. Ann Intern Med 1995, 122:715–724. 24 3 Shamim EA, Rider LG, Miller FW: Update on the genetics of the idiopathic inflammatory myopathies. Curr Opin Rheumatol 2000, 12:482–491. 4 Ollier W: The genetic basis of rheumatic disease. In Rheumatology, edn 3. Edited by Hochberg MC, Silman AJ, Smolen JS, Weinblatt ME, Weisman MH. Mosby; 2003:99–111. Wiendl H, Mitsdoerffer M, Hofmeister V, et al.: The non-classical MHC molecule HLA-G protects human muscle cells from immune-mediated lysis: implications for myoblast transplantation and gene therapy. Brain 2003, 126:176–185. Using in vitro models, the authors studied HLA-G isoforms, and found that they are powerful inhibitors of the primary and secondary immune response in muscle. 5 Rider LG, Gurley RC, Pandey JP, et al.: Clinical, serologic, and immunogenetic features of familial idiopathic inflammatory myopathy. Arthritis Rheum 1998, 41:710–719. 6 Arnett FC, Targoff IN, Mimori T, et al.: Interrelationship of major histocompatibility complex class II alleles and autoantibodies in four ethnic groups with various forms of myositis. Arthritis Rheum 1996, 39:1507–1518. Wiendl H, Behrens L, Maier S, et al.: Muscle fibers in inflammatory myopathies and cultured myoblasts express the nonclassical major histocompatibility antigen HLA-G. Ann Neurol 2000, 48:679–684. 25 • 26 Reed AM: Chimerism in myositis. Curr Rheumatol Rep 2003, 5:421–424. 27 Reed AM, Shock LP, Picornell YJ: Chimerism in children with JDM. Arthritis Rheum 1998, 41:S264. 28 Reed AM, Picornell YJ, Harwood A, Kredich DW: Chimerism in children with juvenile dermatomyositis. Lancet 2000, 356:2156–2157. 7 Hausmanowa-Petrusewicz I, Kowalska-Oledzka E, Miller FW, et al.: Clinical, serologic, and immunogenetic features in Polish patients with idiopathic inflammatory myopathies. Arthritis Rheum 1997, 40:1257–1266. 29 Artlett CM, Ramos R, Jiminez SA, et al.: Chimeric cells of maternal origin in juvenile idiopathic inflammatory myopathies. Childhood Myositis Heterogeneity Collaborative Group. Lancet 2000, 356:2155–2156. 8 Shamim EA, Rider LG, Pandey JP, et al.: Differences in idiopathic inflammatory myopathy phenotypes and genotypes between Mesoamerican Mestizos and North American Caucasians: ethnogeographic influences in the genetics and clinical expression of myositis. Arthritis Rheum 2002, 46:1885–1893. 30 Reed AM, Cragoe S, Kredich D, et al.: Juvenile dermatomyositis and maternal cell chimerism, how does it all begin? Arthritis Rheum 2001, 44:S213. 31 9 Furuya T, Hakoda M, Higami K, et al.: Association of HLA class I and class II alleles with myositis in Japanese patients. J Rheumatol 1998, 25:1109– 1114. Artlett CM, Miller FW, Rider LG: Persistent maternally derived peripheral microchimerism is associated with the juvenile idiopathic inflammatory myopathies. Rheumatology 2001, 40:1279–1284. 32 10 Rider LG, Shamim E, Okada S, et al.: Genetic risk and protective factors for idiopathic inflammatory myopathy in Koreans and American whites: a tale of two loci. Arthritis Rheum 1999, 42:1285–1290. Artlett CM, O’Hanlon TP, Lopez AM, et al.: HLA-DQA1 is not an apparent risk factor for microchimerism in patients with various autoimmune diseases and in healthy individuals. Arthritis Rheum 2003, 48:2567–2572. 11 Rider LG, Artlett CM, Foster CB, et al.: Polymorphisms in the IL-1 receptor antagonist gene VNTR are possible risk factors for juvenile idiopathic inflammatory myopathies. Clin Exp Immunol 2000, 121:47–52. 12 Pachman LM, Liotta-Davis MR, Hong DK, et al.: TNFalpha-308A allele in juvenile dermatomyositis: association with increased production of tumor necrosis factor alpha, disease duration, and pathologic calcifications. Arthritis Rheum 2000, 43:2368–2377. 13 Luppi P, Rossiello MR, Faas S, Trucco M: Genetic background and environment contribute synergistically to the onset of autoimmune diseases. J Mol Med 1995, 73:381–393. 14 Cooper GS, Miller FW, Pandey JP: The role of genetic factors in autoimmune disease: implications for environmental research. Environ Health Perspect 1999, 107(suppl 5):693–700. 15 Hengstman GJ, van Venrooij WJ, Vencovsky J, et al.: The relative prevalence of dermatomyositis and polymyositis in Europe exhibits a latitudinal gradient. Ann Rheum Dis 2000, 59:141–142. Okada S, Weatherhead E, Targoff IN, et al.: Global surface ultraviolet radiation intensity may modulate the clinical and immunologic expression of autoimmune muscle disease. Arthritis Rheum 2003, 48:2285–2293. An important global collaboration from 15 countries looking at 919 IIM patients. The proportion of DM patients, versus all IIM patients, and the proportion of antiMi-2 antibodies, versus all MSAs, were found to correlate with UV intensity. 17 Mierau R, Dick T, Bartz-Bazzanella P, et al.: Strong association of dermatomyositis-specific Mi-2 autoantibodies with a tryptophan at position 9 of the HLA-DR beta chain. Arthritis Rheum 1996, 39:868–876. 18 Hood E: The environment-autoimmune link. Environ Health Perspect 2003, 111:A274–A276. 19 Werth VP, Callen JP, Ang G, Sullivan KE: Associations of tumor necrosis factor alpha and HLA polymorphisms with adult dermatomyositis: implications for a unique pathogenesis. J Invest Dermatol 2002, 119:617–620. 20 Tezak Z, Hoffman EP, Lutz JL, et al.: Gene expression profiling in DQA10501+ children with untreated dermatomyositis: a novel model of pathogenesis. J Immunol 2002, 168:4154–4163. Reed AM, McNallan K, Wettstein P, et al.: Does HLA-dependent chimerism underlie the pathogenesis of juvenile dermatomyositis? J Immunol 2004, 172:5041–5046. The role of maternal microchimeric cells is studied in a large JDM cohort, and for the first time it is demonstrated that chimeric cells in JDM patients are reactive against host cells. 33 •• 34 Lambert NC, Evans PC, Hashizumi TL, et al.: Cutting edge: persistent fetal microchimerism in T lymphocytes is associated with HLA-DQA10501: implications in autoimmunity. J Immunol 2000, 164:5545–5548. 35 Stevens AM: Foreign cells in polymyositis: could stem cell transplantation and pregnancy-derived chimerism lead to the same disease? Curr Rheumatol Rep 2003, 5:437–444. 36 Stevens AM, Sullivan KM, Nelson JL: Polymyositis as a manifestation of chronic graft-versus-host disease. Rheumatol 2003, 42:34–39. •• A fascinating retrospective review of patients undergoing hematopoietic stem cell transplantation over a 30-year period. The incidence of PM was found to be far higher than that of sporadic PM, supporting the alloimmune hypothesis. 37 Nelson JL: Maternal-fetal immunology and autoimmune disease: is some autoimmune disease auto-alloimmune or allo-autoimmune? Arthritis Rheum 1996, 39:191–194. 38 Lampe JB, Gossrau G, Kempe A, et al.: Analysis of HLA class I and II alleles in sporadic inclusion-body myositis. J Neurol 2003, 250:1313–1317. 39 Askanas V, Engel WK: Proposed pathogenetic cascade of inclusion-body myositis: importance of amyloid-beta, misfolded proteins, predisposing genes, and aging. Curr Opin Rheumatol 2003, 15:737–744. 40 Askanas V, McFerrin J, Alvarez RB, et al.: Beta APP gene transfer into cultured human muscle induces inclusion-body myositis aspects. Neuroreport 1997, 8:2155–2158. 16 •• 21 Lutz J, Huwiler KG, Fedczyna T, et al.: Increased plasma thrombospondin-1 (TSP-1) levels are associated with the TNF[alpha]-308A allele in children with juvenile dermatomyositis. Clin Immunol 2002, 103:260–263. Hassan AB, Nikitina-Zake L, Sanjeevi CB, et al.: Association of the proinflammatory haplotype (MICA5.1/TNF2/TNFa2/DRB103) with polymyositis and dermatomyositis. Arthritis Rheum 2004, 50:1013–1015. The authors demonstrate that the ancestral haplotype, together with MICA5.1/TNF2/TNFa2, is an important susceptibility factor for PM/DM. 22 • 23 Wiendl H, Mitsdoerffer M, Weller M: Express and protect yourself: the poten- Askanas V, Engel WK, McFerrin J, Vattemi G: Transthyretin Val122Ile, accumulated Abeta, and inclusion-body myositis aspects in cultured muscle. Neurology 2003, 61:257–260. An elegant gene transfer experiment upregulating the IBM phenotype in muscle fibers of a patient expressing the TTR mutation. 41 • 42 Gossrau G, Gestrich B, Koch R, et al.: Apolipoprotein E and alpha-1antichymotrypsin polymorphisms in sporadic inclusion body myositis. Eur Neurol 2004, 51:215–220. 43 Greenberg SA, Sanoudou D, Haslett JN, et al.: Molecular profiles of inflammatory myopathies. Neurology 2002, 59:1170–1182. 44 Engel AG, Arahata K: Monoclonal antibody analysis of mononuclear cells in myopathies. II: Phenotypes of autoinvasive cells in polymyositis and inclusion body myositis. Ann Neurol 1984, 16:209–215. 45 Hofbauer M, Wiesener S, Babbe H, et al.: Clonal tracking of autoaggressive T cells in polymyositis by combining laser microdissection, single-cell PCR, Disease mechanisms in IIM Chinoy et al. 713 and CDR3-spectratype analysis. Proc Natl Acad Sci USA 2003, 100:4090– 4095. 49 46 Muntzing K, Lindberg C, Moslemi AR, Oldfors A: Inclusion body myositis: clonal expansions of muscle-infiltrating T cells persist over time. Scand J Immunol 2003, 58:195–200. 47 Serreze DV, Pierce MA, Post CM, et al.: Paralytic autoimmune myositis develops in nonobese diabetic mice made Th1 cytokine-deficient by expression of an IFN-gamma receptor beta-chain transgene. J Immunol 2003, 170:2742–2749. 50 Hoffman EP, Brown KJ, Eccleston E: New molecular research technologies in the study of muscle disease. Curr Opin Rheumatol 2003, 15:698–707. • A useful and informative review outlining future approaches in muscle disease investigation, summarizing techniques such as gene expression profiling and whole genome SNP association studies. 48 Nguyen HX, Tidball JG: Expression of a muscle-specific, nitric oxide synthase transgene prevents muscle membrane injury and reduces muscle inflammation during modified muscle use in mice. J Physiol 2003, 550:347–356. van der Meulen MF, Bronner IM, Hoogendijk JE, et al.: Polymyositis: an overdiagnosed entity. Neurology 2003, 61:316–321. 51 Risch N, Merikangas K: The future of genetic studies of complex human diseases. Science 1996, 273:1516–1517. 52 Kruglyak L: Prospects for whole-genome linkage disequilibrium mapping of common disease genes. Nat Genet 1999, 22:139–144. EDITORIAL OVERVIEW Illness and art: the legacy of Paul Klee John Varga Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA Correspondence to John Varga, MD, Division of Rheumatology, Northwestern University Feinberg School of Medicine, McGaw 2300, 240 E. Huron Street, Chicago, IL 60611, USA Tel: 312 503 8003; fax: 312 503 0994, e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:714–717 © 2004 Lippincott Williams & Wilkins 1040–8711 Introduction Along with Pablo Picasso and Henri Matisse, Paul Klee was a dominant figure of 20th century art. His works, characterized by brilliant color, breathtaking inventiveness, spectacular versatility, and unmatched productivity, have had a profound influence on all major graphic artists to follow, and they have fundamentally shaped our sensibility of art. At his untimely death in 1940, Klee left a staggering 10,000 paintings, drawings, and etchings. When he was 56 years old, Klee came down with scleroderma, the illness that was to progressively disable and ultimately kill him. Although its onset had an initially devastating impact, during the remaining 5 years of his life Klee’s struggle with his illness energized him, emboldened his vision, and led him to new insight. It is with his late work that Klee created his artistic manifesto. Klee’s life and art Klee was a complex and enigmatic painter, working with infinite styles and techniques, equally comfortable with abstract and representative art. He was also a gifted musician and composer, a passionate teacher, a poet and philosopher, and an artist who had the courage to stand firmly by his convictions in the face of grave danger. He was a calm self-contained man who spoke little; his art spoke for him. Klee was born in 1879 in Switzerland. His father was a music teacher, and young Klee grew up in a richly cultured and happy home. His childhood was dominated equally by music and art. As a young man he supported himself as a violinist in a chamber orchestra and as a music critic. Except during his final illness, he played the violin every morning for an hour before he got down to work. Throughout his life, Klee’s dedication to, and love for, musical forms informed his drawings and paintings. As reflected in his writings (he left 4000 pages of diary entries, analytical texts, and lecture notes from his years as a teacher), he viewed art through the prism of music. Klee left home at the age of 19 to study painting in Munich. There, he married the concert pianist 714 Lily Stumpf, and their only child, Felix, was born in 1906. Klee’s youthful work is characterized by vitality, a whimsical light touch, and irrepressible humor. He favored prints, etchings, and pen-and-ink drawings that revealed a fondness for the satiric, the grotesque, and the surreal. He gave his pictures evocative and satirical titles such as “Two Men Meet, Each Believing the Other to Be of Higher Rank” (Fig. 1). In 1912, Klee met and exhibited with Vasily Kandinsky and other members of the influential Blaue Reiter Expressionist group. A trip to Tunis in 1914 had an enormous impact on his art. During this trip, Klee experienced North African architecture and intense light; he discovered color. He wrote, “Color has taken possession of me; no longer do I have to chase after it. I know that it has hold of me forever…Color and I are one. I am a painter.” The result was his “square paintings,” which began as squares of sun-splashed colors, often accompanied by triangles or domes, as in “Red and White Domes” (Fig. 2). Although these images are described as abstract, for Klee the meaning of abstraction lay in the opposite direction to the intellectual effort of abstracting. As he famously wrote, “Art does not reproduce the visible; it makes visible.” Although close friends were killed in the World War I, Klee, who was drafted into the German Army at the age of 36, managed to spend two relatively quiet years in a Bavarian garrison, painting airplane wings. With the collapse of Germany, he returned to his family deeply disillusioned by war. In 1925, he was invited to join the faculty of the Bauhaus, the newly established State School of Art and Design in Weimar. During the next 16 years, he taught arts and craft, painting, drawing, bookbinding, stained glass, and textile design. It was a happy and fertile period, with growing complexity of his square paintings and bold experiments with color paralleled by his increasing international reputation. Klee’s 50th birthday was celebrated with a large exhibition in Berlin. With the rise of the Nazis, this tranquil phase of Klee’s life came to an end. Already suspect and accused of being non-Aryan, Klee refused to declare loyalty to the Nazi regime and was dismissed from his job. As his persecution intensified, his works were removed from private and public German collections. In the fall of 1933, Klee decided to leave Germany and settled in his native Bern. In 1937, seventeen of his paintings were included Editorial overview: Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes Varga 715 Figure 1. “Two Men Meet, Each Believing the Other to Be of Higher Rank,” 1903 Etching, black and white. Photographs taken during Klee’s last years attest to the progressive ravages of his illness. The pictures show curling of his fingers, sclerodactyly, hollow cheeks, and taut facies with drawn lips and prominent nose. He appears increasingly thin and is wearing a sweater in his studio, suggesting intolerance to cold. Although no medical records survive, there can be little doubt in retrospect that Klee had scleroderma. Although the “measles” diagnosed at age 56 is difficult to explain (perhaps it was telangiectasia, perceptible in a later photograph of Klee’s face), his exhaustion, stiff hands, dysphagia and weight loss, dyspnea, and ultimately heart failure represent a characteristic clinical picture of progressive scleroderma. Seventy years ago, little could be offered to alleviate the misery of scleroderma or to halt its progression. in Joseph Goebbels’ infamous “Entartete Kunst” (Degenerate Art) show. The exhibition, including also works by Picasso, Edvard Munch, Marc Chagall, Oskar Kokoschka, and others, was designed to ridicule and denigrate creative art that did not uphold “correct” National Socialist virtues, and was seen by millions of Germans. Two years later, much of the “Degenerate Art” was burned in the courtyard of a Berlin fire station, and others were auctioned off to the highest bidders. Although in poor health, and shaken by the recent death of his father, in 1940 Klee mounted a final exhibition in Zurich, featuring more than 300 pieces. The exhibition was badly received, with negative allusions to the artist’s mental state. Exhausted, he entered a sanitarium for several weeks. In May 1940, feeling that the end was near, Klee was accompanied by his wife to a nursing home in Locarno. In order not to upset him, no one talked about the war or the fall of Paris. He died on the morning of June 29. His death was attributed to heart failure. Klee’s flight and illness The impact of illness on Klee’s art The flight from Germany had shaken Klee severely; he lost his home, his professorship, his culture. Although his name was already famous around the world, few people in his native town had heard of him. Worse, all the money he had earned and banked in Germany was lost; he was back once more in the financial state he had been when he first left Bern 30 years before. Ironically, he never became a citizen of Switzerland, the country of his birth; his application, initially turned down by the authorities, was finally granted only after his death. How did his illness influence Klee’s art, productivity, creativity, composition, and technique? The numbers are telling. Klee, who was Germanic in his passion for cata- On top of his isolation in his new surroundings, Klee started to feel exhausted. Although he was characteristically stoic and kept his concerns largely to himself, his wife made reference to his fatigue in her correspondence. In 1935, Klee experienced a skin rash that was diagnosed as measles. According to his son, he never fully recovered, and the “measles” was followed by a succession of illnesses. He had difficulty swallowing, and eventually he could only take a liquid diet. Because of his embarrassment, he ate his meals alone. An avid hiker, he experienced exertional shortness of breath. In his letters, he described having arthritic pain in his hands, making it difficult to hold the paintbrush. He visited several doctors and health spas. Finally, the diagnosis of scleroderma was made in 1936. As his health declined his doctors ordered him to stop smoking. Around this time, he had also to give up playing the violin. His friends Picasso and Georges Braque came to pay homage. Figure 2. “ Red and White Domes,” 1914 Watercolor. 716 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes loguing his artistic output, recorded that in 1936, after the onset of his illness, he produced only 25 works. This was a dramatic fall in productivity. However, subsequent years saw a progressive increase, with a staggering 1253 pieces completed in 1939, the last full year of his life! It is as if the struggle to come to grips with his mortality allowed Klee to draw on enormous stores of creative energy, imagination, and power. And what about the nature of his later work? A gradual but profound evolution took place. These changes are marked by greater simplicity and greater intensity. Klee began to use coarser mediums like poster paints, and rough materials such as burlap and newspaper. He abandoned his intricate, small-scale compositions for much larger pieces, some longer than 2 meters. The late pictures are bolder; abstract figures are painted with simple, heavy black lines; some seem childish or primitive. The lightheartedness and wit of the early works now gives way to introspection and despair. The mood is somber, the colors are dull, the subjects become symbols; gone are the whimsy and buoyancy, the exuberant yellows, greens, and blues, of 20 years before. The titles (“Forgetful Angel,” “Gloomy Cruise,” “Hurt,” “The Sick One in the Boat”) reflect the themes of suffering, destruction, and death, signifying Klee’s anguish over his illness and no doubt also his gloom over the inevitability of another world war. Figures of angels and devils betray the artist’s fear of death. As Gunter Wolf noted, in many late pictures we see the reflection of the profound changes in Klee’s own face and body. “Maske” (The Mask, 1940) and “Durchhalten!” (Endure! 1940) show a disfigured face resembling the artist. Bars of thick brushstrokes, menacing and emblematic, make their appearance, often with a skeletal figure behind a grid of bars. Art historians have puzzled over the meaning of the recurrent bars; but it is not hard to look at “The Captive” (Fig. 3) and see the artist trapped in the prison of scleroderma, the steel cage of his own physical immobility. Like his favorite composer, Mozart, Klee created his own disturbing requiem, “Tod und Feuer” (Death and Fire, 1940). The painting, one of his last, is dominated by a gleaming white skull, with the word “Tod” (Death) forming the features of the face. A solitary stick figure walks toward the skull; deep shades of red and orange signify burning, a combustion, in contrast to a cool graygreen domain of death below. The featureless man with a body of no substance (Klee?) walks forward without hesitation, even though his next step is into his own grave. Klee now knew that the end was approaching. He was now unafraid of death: requiem, not despair. Klee’s body was cremated, and his ashes were interred in Berne. On his grave are inscribed these words from his diary: Figure 3. “The Captive,” 1940 Oil on burlap. I cannot be grasped in the here and now For I live as well with the dead As with the yet unborn. A little nearer to the heart of creation than is normal But still not close enough. Klee’s legacy Klee’s artistic output was staggering, and his legacy as a writer and philosopher equally copious. He worked in all mediums and moved freely between abstraction and representation. Unlike other graphic artists, Klee’s reputation is not tied to a single, easily identifiable masterpiece; rather, his approach to art was oriented toward a lifelong process of reexamining and redefining themes and form. His career was one of ceaseless experimentation with new techniques, styles, and colors. His creative genius remained undiminished throughout his life and was fundamentally shaped by his final illness. After a period of despair following his flight from Germany and the onset of scleroderma, during which he almost ceased artistic activity altogether, he showed a remarkable turnabout. Even as his health deteriorated and his physical energy declined, his art and creativity mysteriously soared. This paradox ultimately remains Klee’s personal triumph and lasting legacy. Editorial overview: Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes Varga 717 References and recommended reading Bywater EGL: Paul Klee: the effect of scleroderma on his painting. Rheumatic Disease in Visual Arts. 1997: 49–50. Papers of particular interest, published within the annual period of review, have been highlighted as: Grohmann W. Klee. New York: Harry N. Abrams, 1985. • Of special interest •• Of outstanding interest Bridget Riley, Hayward Gallery Exhibit, February 2002. Silver R: Paul Klee and scleroderma. Bull Rheum Dis 1995, 45:4–6. Klee: Gualtieri di San Lazzarro. New York: Praeger, 1964. Paul Klee at the Guggenheim Museum. New York: Guggenheim Museum, 1993. The diaries of Paul Klee: Edited by Felix Klee. Berkeley: University of California Press, 1964. Keller C, Dotz W, Varga J: Scleroderma of Paul Klee. Dermatopathology 1999, 5:19–22. Wolf G: Endure!: how Paul Klee’s illness influenced his art. Lancet 1999, 353:1516–1518. Raynaud phenomenon and the vascular disease in scleroderma M. Bashar Kahaleh Purpose of review Raynaud phenomenon is the earliest and most common clinical manifestations of scleroderma (systemic sclerosis). Therefore, Raynaud phenomenon offers the best window into the investigation of the early steps in the pathogenesis of systemic sclerosis. This review focuses on the differential diagnosis of Raynaud phenomenon, the transition of Raynaud phenomenon to systemic sclerosis, mechanisms and consequences of vascular injury and dysfunction in systemic sclerosis, and therapeutic options. Recent findings Careful clinical evaluation using a simple definition of Raynaud phenomenon is the most reliable and reproducible method in the diagnosis. Although the assessment of vascular function by noninvasive methods is still not sensitive enough for the evaluation and follow-up of individual patients, it helps in the differential diagnosis and in population studies. Progressive deficiency in vasodilatory capacity of the vessels is proposed as a mechanism of Raynaud phenomenon, particularly in systemic sclerosis. In addition, decreased fibrinolysis and enhanced coagulation pathways undoubtedly contribute to vascular dysfunction. The mechanism of endothelial injury is still elusive, yet endothelial apoptosis mediated by antiendothelial antibodies is the most attractive hypothesis now. Therapies directed at the vascular disease continue to focus on the alleviation of vascular spasm. However, immunosuppressive therapy may influence the levels of vascular injury markers and thus may have an effect on the vascular disease itself. Summary Continued progress in the investigation of the vascular aspects of scleroderma is described in this review. Immune involvement in the early stages of the disease and mechanism of vascular repair in advance cases are some of the highlights of last year’s progress. Keywords Raynaud phenomenon, endothelial cells, vascular disease Curr Opin Rheumatol 16:718–722. © 2004 Lippincott Williams & Wilkins. Division of Rheumatology, Department of Medicine, Medical College of Ohio, Toledo, Ohio, USA Correspondence to M. Bashar Kahaleh, MD, Professor of Medicine, Chief, Division of Rheumatology, Medical College of Ohio, 1321 Glendale Avenue, Toledo, OH 43617, USA Tel: 419 383 4271; fax: 419 383 6244; e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:718–722 718 Abbreviation CTD connective tissue disease © 2004 Lippincott Williams & Wilkins 1040–8711 Introduction Raynaud phenomenon is an exceedingly important component of systemic sclerosis because it symbolizes the generalized and progressive nature of the vascular dysfunction in the disease. The histopathologic bases for the vascular dysfunction are well described and involve signs of injury to the microcirculation and small blood vessels. A core set variables for the assessment of vascular involvement in systemic sclerosis was proposed [1]. Raynaud phenomenon and digital ulcers were identified as the two parameters that represent the minimal requirements for the documentation of vascular involvement in systemic sclerosis. Attacks of Raynaud phenomenon should be characterized by duration, frequency, severity, and the Raynaud condition score. The assessment of digital ulcers should include frequency, size, severity, number of involved digits, and the VAS score. Relevant studies published last year are reviewed and grouped in sections related to the transition of Raynaud phenomenon to connective tissue disease, the noninvasive evaluation of primary and secondary Raynaud phenomenon, etiologic factors in Raynaud phenomenon, and vascular disease and proposed therapeutic options. Transition to connective tissue disease One of the most difficult clinical challenges faced when evaluating patients with Raynaud phenomenon is the assessment of risk for transition to connective tissue disease (CTD). DeAngelis et al. [3] reported the outcome of 118 consecutive patients with Raynaud phenomenon attending a rheumatology specialty clinic. Thirty-five patients were classified as having primary Raynaud phenomenon, 20 were unclassifiable, and 63 patients had secondary Raynaud phenomenon. After an average follow-up of 3 years, none of the primary group developed secondary condition, and only 10% of the unclassifiable group developed systemic sclerosis. This report documents the rare transition from primary Raynaud phenomenon to CTD even in patients with suspected CTD. In contrast, Ziegler et al. [4•] reported a 9% tran- Raynaud phenomenon and the vascular disease in scleroderma Kahaleh 719 sition from primary Raynaud phenomenon and 30% from possible secondary Raynaud phenomenon after a 12.4year mean follow-up. This study suggests that there is a continuum in transition to CTD and that clinicians should not confidently assure patients with primary Raynaud phenomenon of the benign nature of their symptoms. as in the carotid arteries. Duplex determination of the carotid elasticity or stiffness was significantly different in the two groups [10]. The same investigators reported in a subsequent study more reduction in elastic properties of the carotid artery in the dSSc subset than in lSSc [11]. The two studies document the frequent, but often neglected, large vessel involvement in systemic sclerosis. The value of positive ANA in predicting future development of CTD was examined by Myckatyn and Russell [5], who described the outcome of patients with positive ANA using the clinical database at the University of Alberta Hospital. After a mean follow-up of 5.4 years, 91% remained ANA-positive, and 6% progressed to CTD. The study emphasized the long-term persistence of positive serology and the low rate of transition to CTD. Etiologic and pathogenetic factors in Raynaud phenomenon and systemic sclerosis vascular disease Genetic factors Childhood Raynaud phenomenon is rare and not well characterized. One of the largest series of children with Raynaud phenomenon was elegantly described [2•]. The mean age at onset was 12 years (range, 1–19 years). Seventy percent were classified as having primary Raynaud phenomenon, and 30% were associated with other diseases. One of the surprising findings in this study was the presence of antiphospholipid antibodies in more than 20% of children with Raynaud phenomenon. Evaluation of Raynaud phenomenon The diagnostic value of nailfold capillaroscopy was evaluated in 447 patients with CTD. The results again confirmed the specificity of systemic sclerosis capillary pattern [6]. A comparison of thermography and laser Doppler imaging in the assessment of Raynaud phenomenon demonstrated poor correlation between the two methods. The lack of correlation suggests that one technique is not a good substitute for the other [7]. The authors suggested that Doppler imaging is more sensitive to changes than thermography. Ultrasound examination of the microcirculation was performed with newly developed multi-D linear array transducer that improves resolution [8]. Thirty-three patients, 14 with primary Raynaud phenomenon and 19 with systemic sclerosis, were examined. Differences in the diameters of the digital arteries allowed the differentiation of primary Raynaud phenomenon from systemic sclerosisassociated Raynaud phenomenon. Primary Raynaud phenomenon can also be distinguished from systemic sclerosis-associated Raynaud phenomenon by the assessment of skin perfusion pressure at rest and after cold presser [9]. Significant reduction in perfusion pressure was noted in the systemic sclerosis group that may be related to the obstructive arteriolar lesion in systemic sclerosis microcirculation. Differences between primary and secondary Raynaud phenomenon can also be shown in the large circulation, The link of genetic factors to Raynaud phenomenon was suggested in a case report describing primary Raynaud phenomenon in 16-year-old monozygotic twins [12]. The concordance rate for Raynaud phenomenon in monozygotic twins is unknown. Nonetheless, significant familial aggregation of primary Raynaud phenomenon is well described. Investigation of large series of twins and multicase families is needed to explore the role of genetics in the pathogenesis of Raynaud phenomenon. Endothelial-dependent relaxation Defective endothelial dependent vasodilatation in systemic sclerosis is well described. Schlez et al. [22] extended these observations by evaluating blood flow velocity and the diameter of skin capillaries before and after the microinjection of acetylcholine (endothelialdependent) or sodium nitroprusside (endothelial-independent) in 10 systemic sclerosis and control subjects. No difference in respond to nitroprusside was noted; however, significant defect in response to acetylcholine was noted in patients with systemic sclerosis. Moreover, the response to acetylcholine returned to normal in two patients with systemic sclerosis after 1 week of prostacyclin infusion. Although this is a limited observation, the potential for prostacyclin as a vascular modifying agent in systemic sclerosis has been long suggested. Coagulation and fibrinolysis The status of the coagulation and fibrinolytic systems in systemic sclerosis is not well described. Mattuci-Cerinic et al. [13•] examined the coagulation/fibrinolysis system in 29 patients with systemic sclerosis. Significant activation of the coagulation system and reduction in fibrinolysis activities was noted. The conclusions in this study are in agreement with the histologic findings in systemic sclerosis, in which excessive fibrin deposits are commonly seen in association with thrombosis in the microvessels. Still, no controlled trial using anticoagulation or fibrinolytic enhancing therapy in systemic sclerosis has been accomplished to date. Atherosclerosis and hyperlipidemia van Vugt et al. [14] reported a higher than expected incidence of arteriosclerotic disease and hyperlipidemia in patients with primary Raynaud phenomenon who under- 720 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes went upper extremity angiograms. Of 103 patients with primary Raynaud phenomenon, the angiograms were compatible with vasospasm in 42 patients, atherosclerotic vascular disease in 44 patients, peripheral embolism in eight patients, and vasculitis and Buerger disease in six patients. Moreover, 47% of patients had hyperlipidemia. The high frequency of atherosclerotic vascular disease in patients with Raynaud phenomenon noted in this study should serve as a reminder of the epidemic nature of atherosclerosis and should encourage the search for hyperlipidemia in patients with Raynaud phenomenon. Endothelial apoptosis The bulk of published studies suggest a pathogenetic role for antiendothelial antibodies in systemic sclerosis. This conclusion was reinforced by Worda et al. [15••], who described the induction of endothelial apoptosis in the chicken model of systemic sclerosis (UCD-200) by directly transferring UCD-200 serum into normal chicken embryos. Binding of antiendothelial antibodies to the microvascular EC in the chorioallantoic membrane in association with EC apoptosis was unmistakably seen. However, the cell and target antigen specificity of antiendothelial antibodies is still unknown. Moreover, standarized detection methods are badly needed. A new assay for antiendothelial antibodies that used indirect immunofluorescence technique with rodent lung preparations and fluorescinated human anti-IgG has been introduced [16]. Positive testing was recorded in 42 of 45 patients with systemic sclerosis. It is not clear whether this method detects ANAs in addition to antiendothelial antibodies, because the reported staining patterns were nuclear or nucleolar in location. Moreover, it is not clear whether antiendothelial antibodies are truly endothelialspecific and whether they react with the same antigens. Indeed, Western blot analysis of EC protein extracts showed an average of 10 reacting bands in each antiendothelial antibody-positive serum, and in most examples, extracts from fibroblast reacted with antiendothelial antibodies, sometimes even more so than with EC. Vasculogenesis and angiogenesis The vascular changes in systemic sclerosis affect predominantly the microcirculation and arterioles, with capillary necrosis and intimal proliferation of arterioles resulting in occlusion of blood vessels, decreased organ blood flow, and a state of progressive chronic organ ischemia. This underperfused state (ischemia, acidosis) should be a fertile ground for neoangiogenesis, but new capillaries are rare, and broad avascular areas are common, suggesting defective pathways in angiogenesis and vascular repair. Konttinen et al. [17•] reported an immunohistochemical study of systemic sclerosis skin that showed rare and sporadic expression of ␣v3 integrin in the blood vessels, suggesting unsuccessful attempts at new vessel formation. The expression of ␣v3 integrin receptor is associated with VEGF-mediated angiogenesis and is used as a histologic marker for new vessel formation. Circulating endothelial cells The recent innovations in cell isolation and immunohistochemical evaluation allowed the isolation of circulating fully differentiated EC and bone marrow-derived endothelial progenitor cells. Del Papa et al. [18••] observed increased circulating EC and bone marrow-derived endothelial progenitors in 46 patients with systemic sclerosis. The circulating number of ECs correlated with the overall disease activity score and with pulmonary hypertension. Gene expression profile in systemic sclerosis skin Analysis of general and cell type-specific gene expression patterns in systemic sclerosis skin demonstrated upregulation of genes related to endothelial cells (VEcadherin, Thy 1, von Wilbrand factor, and CD31), B and T cells, and extracellular matrix [19••]. The upregulated genes were detected in involved and clinically uninvolved systemic sclerosis skin. It is interesting to note that histologic evidence for fibrosis was seen in clinically unaffected skin. Moreover, no clear differences in patterns of gene expression among fibroblasts derived from systemic sclerosis, morphia, and normal skin were noted, suggesting that fibroblasts in vitro are not a suitable starting sample for examining systemic sclerosis-specific gene expression patterns. Immune involvement Lymphocyte transendothelial migration Enhanced expression of adhesion molecules in systemic sclerosis vessels is widely documented and presumed to result in inflammatory cell trafficking in the vessel wall and surrounding tissue. The role of lymphocyte in this process was examined by Stummvoll et al. [20], who investigated the transendothelial migrating capacity of control and systemic sclerosis peripheral lymphocytes in an in vitro system. Increased migration of systemic sclerosis cells was seen mainly by the CD3+ CD4+ cell subset. Among migrating systemic sclerosis CD4+ T lymphocytes, the frequency of HLA-DR+ cells was increased, and cells were in an activated state, as reflected by enhanced expression of the adhesion molecules CD11a, CD49d, CD29, and CD44. It is important to note that this cell type is frequently seen in the perivascular spaces in the early stages of systemic sclerosis. Cytokines An increased circulating level of interleukin-13 (a cytokine that promotes fibrosis and inflammation) in systemic sclerosis was described in 1997. A study by Riccieri et al. [21] confirmed this observation and correlated circulating levels with parameters of microvascular injury Raynaud phenomenon and the vascular disease in scleroderma Kahaleh 721 assessed by nailfold capillaroscopy. Thus, interleukin-13 levels correlated with an active capillary pattern, the presence of hemorrhages, sludging of blood, and larger total loops and arterial diameters. This observation hints at a possible vascular effect for this cytokine. Therapeutic interventions Therapy of the vascular disease in systemic sclerosis is directed mostly at alleviation of vasospasm. Sildenafil A case report described improved digital perfusion and reduced frequency and severity of Raynaud attacks after sildenafil treatment [23]. effect for L-arginine in primary and secondary Raynaud phenomenon. Ascorbic acid The hypothesis that a potent water-soluble antioxidant can reverse endothelial dysfunction was tested in 11 patients with systemic sclerosis and 10 healthy subjects [28]. The study was a double-blind, randomized, crossover, placebo-controlled trial using 2 g ascorbic acid or placebo. No clear effect for ascorbic acid on endothelialdependent vasodilatation was seen. The authors suggested that the use of different antioxidants or different dosing of ascorbic acid may be required to show beneficial effects for antioxidants on endothelial vasodilatory function. PGE1 Similar findings were noted using transdermal PGE1 ethylester [24]. This study showed improved skin perfusion and reduced frequency of Raynaud phenomenon attacks after the transdermal PGE1 treatment (10 hours daily for 2 weeks). Iloprost The efficacy and safety of intravenous iloprost in the treatment of ischemic digits in pediatric patients with connective tissue diseases who failed to respond to conservative treatment was reported in a retrospective study [25]. Healing of ischemic ulcer and improved blood flow were noted in most patients. Cilostazol Cilostazol (Pletal), a synthetic phosphodiesterase III inhibitor that reversibly inhibits platelet aggregation and is used for the treatment of intermittent lower extremity claudication, was tested in a 6-week, double-blind, placebo-controlled trial of patients with primary and secondary Raynaud phenomenon. Treatment was associated with vasodilation of the brachial arteries and conduit vessels [26]. However, the drug had no effects on microvascular blood flow or on the frequency and severity of Raynaud phenomenon attacks in both primary and secondary Raynaud phenomenon. Still, a multicenter control trial is currently underway to evaluate the effect of cilostazol in children with primary and secondary Raynaud phenomenon (http://www.ClinicalTrials.gov). Cyclophosphamide and prednisone The circulating levels of the endothelial injury markers E-selectin and thrombomodulin were examined in patients with early diffuse systemic sclerosis after 1-year therapy with oral cyclophosphamide and prednisolone [29]. Significant reduction of the circulating levels was noted in association with improved skin scores and pulmonary function at the end of the follow-up period. This intriguing observation suggests a possible role for immunosuppression in the therapy of systemic sclerosis vascular disease. Conclusion Vascular disease is the leading cause of complications and mortalities in systemic sclerosis. Humeral and cellular immunity is important in the development of the early events of vascular disease. Failure of vascular repair in established disease is highlighted. Unfortunately, medical therapy still focuses on the elevation of vasospasm. Better understanding of the mechanisms of endothelial injury and vascular dysfunction will undoubtedly lead to better therapeutic approaches to the vascular aspects of systemic sclerosis. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest 1 L-arginine Defective EDRF (nitric oxide) pathway in Raynaud phenomenon has been suggested, but it has been difficult to overcome therapeutically. L-arginine supplementation has been suggested as a potentially successful strategy. Success in reversing digital ischemia was reported in two patients, and significant improvement in symptoms of Raynaud phenomenon was noted in an additional two patients [27]. L-arginine may be considered for the acute management of digital ischemia; however, numerous previous studies failed to show a long-term Kahaleh B, Meyer O, Scorza R: Assessment of vascular involvement. Clin Exp Rheumatol 2003, 21:S9–14. 2 Nigrovic PA, Fuhlbrigge RC, Sundel RP: Raynaud’s phenomenon in children: a retrospective review of 123 patients. Pediatrics 2003, 111:715–721. • Well-described large series of children with Raynaud phenomenon. Girls were more affected than boys; the majority of cases were primary Raynaud phenomenon. Positive ANA and nailfold capillary pattern correlated with secondary causes. Surprisingly, antiphospholipid antibodies were found in > 20% of children with Raynaud phenomenon. 3 DeAngelis R, Del Medico R, Blasetti P, et al.: Raynaud’s phenomenon: clinical spectrum of 118 patients. Clin Rheumatol 2003, 22:279–284. Ziegler S, Brunner M, Eigenbauer E, et al.: Long-term outcome of primary Raynaud’s phenomenon and its conversion to connective tissue disease: a 12-year retrospective patient analysis. Scand J Rheumatol 2003, 32:343– 347. The frequency of connective tissue disease development in patients considered to 4 • 722 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes have idiopathic Raynaud phenomenon for > 10 years was examined. Overall, 14% progressed to a definite CTD, 10 from primary Raynaud phenomenon and 10 from possible secondary Raynaud phenomenon. The initial presence of antinuclear antibodies, thickening of fingers, higher age at onset of Raynaud phenomenon, and female sex seemed to be important determinants for a possible transition to a CTD. 5 Myckatyn SO, Russell AS: Outcome of positive antinuclear antibodies in individuals without connective tissue disease. J Rheumatol 2003, 30:736– 739. 6 Nagy Z, Czirjak L: Nailfold digital capillaroscopy in 447 patients with connective tissue disease and Raynaud’s disease. J Eur Acad Dermatol Venereol 2004, 18:62–68. 7 Clark S, Dunn G, Moore T, et al.: Comparison of thermography and laser Doppler imaging in the assessment of Raynaud’s phenomenon. Microvasc Res 2003, 66:73–76. 8 Cazalets C, Cador B, Rolland Y, et al.: Digital flow exploration by color Doppler ultrasound in patients with Raynaud’s disease or systemic sclerosis. J Mal Vasc 2004, 29:12–20. 9 Kanetaka T, Komiyama T, Onozuka A, et al.: Laser Doppler skin perfusion pressure in the assessment of Raynaud’s phenomenon. Eur J Vasc Endovasc Surg 2004, 27:414–416. Del Papa N, Colombo G, Fracchiolla N, et al.: Circulating endothelial cells as a marker of ongoing vascular disease in systemic sclerosis. Arthritis Rheum 2004, 50:1296–1304. Not long ago, new vessel formation was believed to be caused by expansion of the existing vascular tree by angiogenesis. Now it is known that bone marrow-derived endothelial progenitors mediate vasculogenesis, and these cells have been shown to be of enormous therapeutic potential in the management of ischemic disorders. This study showed an increase in endothelial progenitors and fully differentiated endothelial cells in systemic sclerosis circulation. Still, new vessel formation is uncommon in systemic sclerosis. Examination of steps in vasculogenesis in systemic sclerosis is clearly needed. 18 •• Whitfield ML, Finlay DR, Murray JI, et al.: Systemic and cel type-specific gene expression patterns in scleroderma skin. Proc Natl Acad Sci USA 2003, 100:12319–12324. Large-scale gene expression study that showed what appears to be consistent differences in gene expression between systemic sclerosis and control skin biopsies irrespective of the clinical appearance of the skin at the biopsy site, suggesting a systemic nature of disease. Moreover, no obvious differences in patterns of gene expression among fibroblasts derived from systemic sclerosis, morphia, and normal skin were noted. 19 •• 20 Stummvoll GH, Aringer M, Grisar J, et al.: Increased transendothelial migration of scleroderma lymphocytes. Ann Rheum Dis 2004, 63:569–574. 10 Cheng KS, Tiwari A, Boutin A, et al.: Differentiation of primary and secondary Raynaud’s disease by carotid arterial stiffness. Eur J Vasc Endovasc Surg 2003, 25:336–341. 21 Riccieri V, Rinaldi T, Spadaro A, et al.: Interleukin-13 in systemic sclerosis: relationship to nailfold capillaroscopy abnormalities. Clin Rheumatol 2003, 22:102–106. 11 Cheng KS, Tiwari A, Boutin A, et al.: Carotid and femoral arterial wall mechanics in scleroderma. Rheumatology (Oxford) 2003, 42:1299–1305. 22 Schlez A, Kittel M, Braun S, et al.: Endothelium-dependent regulation of cutaneous microcirculation in patients with systemic scleroderma. J Invest Dermatol 2003, 120:332–334. 23 Rosenkratz S, Diet F, Weibrauch J, et al.: Sildenafil improved pulmonary hypertension and peripheral blood flow in a patient with sclerodermaassociated lung fibrosis and the Raynaud phenomenon. Ann Med 2003, 139:871–873. 24 Schuzl A, Hufner HM, Kittel M, et al.: Systemic scleroderma patients have improved skin perfusion after the transdermal application of PGE1 ethyl ester. Vasa 2003, 32:83–86. 25 Zulian F, Corona F, Gerloni V, et al.: Safety and efficacy of iolprost for the treatment of ischaemic digits in paediatric connective tissue. Rheumatology (Oxford) 2003, 43:229–233. 26 Rajagopalan S, Pfenninger D, Somers E, et al.: Effects of cilostazol in patients with Raynaud’s syndrome. Am J Cardiol 2003, 92:1310–1315. 27 Rembold CM, Ayers CR: Oral L-arginine can reverse digital necrosis in Raynaud’s phenomenon. Mol Cell Biochem 2003, 244:139–141. 28 Mavrikakis ME, Lekakis JP, Papamichael CM, et al.: Ascorbic acid does not improve endothelium-dependent flow-mediated dilatation of the brachial artery in patients with Raynaud’s phenomenon secondary to systemic sclerosis. Int J Vitam Nutr Res 2003, 73:3–7. 29 Apras S, Ertenli I, Ozbalkan Z, et al.: Effects of oral cyclophosphamide and prednisolone therapy on the endothelial functions and clinical findings in patients with early diffuse systemic sclerosis. Arthritis Rheum 2003, 48:2256– 2261. 12 Oskay T, Olmez U: Primary Raynaud’s phenomenon in monozygotic twins. Ann Rheum Dis 2004, 63:219. Mattuci-Cerinic M, Valentinie G, Sorano GG, et al.: Blood coagulation, fibrinolysis, and markers of endothelial dysfunction in systemic sclerosis. Semin Arthritis Rheum 2003, 32:285–295. Comprehensive study that examined coagulation and fibrinolysis parameters and levels of endothelial injury markers in patients with systemic sclerosis. The results clearly demonstrate reduced fibrinolytic potential and increased coagulation pathway markers along with increased markers of vascular injury. 13 • 14 van Vugt RM, Kater L, Dijkstra PF, et al.: The outcome of angiography in patients with Raynaud’s phenomenon: an unexpected role for atherosclerosis and hypercholesterolemia. Clin Exp Rheumatol 2003, 21:445–450. Worda M, Sgonc R, Dietrich H, et al.: In vivo analysis of the apoptosisinducing effect of anti-endothelial cell antibodies in systemic sclerosis by the chorionallantoic membrane assay. Arthritis Rheum 2003, 48:2605–2614. This excellent study is the first to demonstrate the in vivo apoptosis-inducing effects of antiendothelial antibodies. These findings support a primary pathogenetic role for the antibodies in systemic sclerosis. 15 •• 16 Wusirika R, Ferri C, Marin M, et al.: The assessment of anti-endothelial cell antibodies in scleroderma-associated pulmonary fibrosis. Am J Clin Pathol 2003, 120:596–606. 17 Konttinen YT, Mackiewicz Z, Ruuttila P, et al.: Vascular damage and lack of angiogenesis in systemic sclerosis skin. Clin Rheumatol 2003, 22:196–202. • Although some patients with systemic sclerosis showed a high degree of ␣v3 integrin receptor expression, the majority did not, suggesting defective angiogenesis potentials in systemic sclerosis. Autoantibodies in systemic sclerosis and fibrosing syndromes: clinical indications and relevance Eduardo J. Cepeda and John D. Reveille Purpose of review Systemic sclerosis, or scleroderma, is associated with a variety of autoantibodies, each of them having their own clinical associations. The fibrosing disorders, other than systemic sclerosis, represent a diverse group of diseases with systemic or localized effect and with limited understanding of their pathogenesis. The purpose of this review is to analyze the literature on the clinical usefulness of examining serum autoantibodies in patients with known or suspected scleroderma and fibrosing disorders. Recent findings Studies on autoantibodies within the past year highlight their clinical utility in systemic sclerosis. Anticentromere antibodies are most often seen with limited cutaneous involvement and lower frequency of pulmonary fibrosis and lower mortality (despite an increased risk for pulmonary hypertension) compared with anti-Scl-70 and antinucleolar antibodies. Anti-Scl-70 antibodies are associated with diffuse cutaneous involvement, increased frequency of pulmonary fibrosis, and higher mortality. The anti-polymyositis-scleroderma autoantibody is associated with the polymyositis-scleroderma overlap syndrome. Anti-Th/To antibodies are associated with milder skin and systemic involvement but with more severe pulmonary fibrosis and overall worse prognosis. Anti-RNA-polymerase family antibodies and antifibrillarin antibodies are predictive of diffuse cutaneous and systemic involvement and greater mortality. Less specific autoantibodies for systemic sclerosis and limited data on some other autoantibodies limit their clinical utility in patients with systemic sclerosis. For the most part, the association between autoantibodies and fibrosing disorders other than systemic sclerosis remains inconclusive. Summary Autoantibodies in systemic sclerosis provide important and prognostic information and are useful in defining clinical subsets of the disease. When used appropriately, they can be a useful instrument in the management of scleroderma. Keywords autoantibodies, systemic sclerosis, Raynaud phenomenon, overlap syndrome, fibrosing disorders Curr Opin Rheumatol 16:723–732. © 2004 Lippincott Williams & Wilkins. Division of Rheumatology, The University of Texas-Houston Health Science Center at Houston, Houston, Texas, USA Correspondence to John D. Reveille, The University of Texas-Houston Health Science Center, MSB 5.270, 6431 Fannin, Houston, TX 77030, USA Tel: 713 500 6900; fax: 713 500 0580; e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:723–732 Abbreviations ACA aCL AFA ANoA aPL ELISA LAC MHC RNAP SLE SSc TNF anticentromere antibodies anticardiolipin antibodies antifibrillarin antibodies antinucleolar antibodies antiphospholipid antibodies enzyme-linked immunosorbent assay lupus anticoagulant major histocompatibility complex RNA polymerase systemic lupus erythematosus systemic sclerosis, scleroderma tumor necrosis factor © 2004 Lippincott Williams & Wilkins 1040–8711 Introduction Patients with systemic sclerosis (scleroderma, SSc) express a variety of autoantibodies that have their own clinical associations. Whether these autoantibodies play a direct role in the pathogenesis of SSc or are merely epiphenomena, they carry significant value in both diagnosis and prognosis. Moreover, autoantibodies in SSc are clearly influenced by hereditary factors, as evidenced by their higher concordance in identical twins and their associations with major histocompatibility complex (MHC) genes [1•,2]. The autoantibodies classically associated with SSc include anticentromere antibodies (ACA) and anti-Scl-70 (anti-topoisomerase-I). In addition to these are the less commonly occurring components of the antinucleolar antibody (ANoA) system, which comprises a mutually exclusive heterogeneous group of autoantibodies that produce nucleolar staining by indirect immunofluorescence on cells from a variety of species [3]. The most widely recognized of these include anti-PM-Scl, antifibrillarin/anti-U3-ribonucloprotein (AFA), anti-Th/To, and the anti-RNA-polymerase family (anti-RNAP), including anti-RNAP I, II, and III (although anti-RNAP frequently do not produce nucleolar staining on immunifluorescence) [4–9]. In addition to these disease-specific antibodies, other autoantibodies are also found in patients with SSc, with varying clinical significance. The purpose of this review is to describe the pathogenic significance and clinical utility of determining various 723 724 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes autoantibodies associated with SSc, Raynaud phenomenon, overlap syndromes with SSc, and fibrosing disorders. one recent study, ACA positivity correlated with longerduration Raynaud phenomenon before the diagnosis of SSc was made [17]. Anticentromere antibodies Anticentromere antibodies are most often seen in the presence of limited cutaneous SSc and are associated with a higher risk for calcinosis and ischemic digital loss in patients with SSc [10••,18–21,22•,23]. Likewise, there is a significantly lower frequency of interstitial pulmonary fibrosis in SSc with ACA [10••,20,21]. Anticentromere antibodies have been most typically determined by their characteristic staining pattern on immunofluorescence, giving rise to a speckled appearance on Hep-2 cells. More recent techniques, including enzyme-linked immunosorbent assay (ELISA) and immunoblotting have been developed and are being used increasingly in clinical practice. They have been shown to perform with satisfactory accuracy when compared with indirect immunofluorescence [10••]. Thus far, six centromeric nucleoproteins, CENP-A through CENP-F, are known to be bound by sera from patients with SSc [10••]. However, these distinctions have not been shown to have clinical relevance. All sera containing ACA react with CENP-B. A solid-phase ELISA has been established by using a cloned fusion protein of CENP-B as antigen. More recently, this ELISA has been refined and has been found to have adequate sensitivity and specificity for clinical use (Table 1) [10••]. The frequency of ACA in patients with SSc has been reported to be 20 to 30% overall, but it varies in different ethnic groups. They occur most frequently in Caucasians, where they are found in approximately a third of SSc patients, compared with a significantly lower frequency in Hispanic, African-American, and Thai patients [11,12,13•]. ACA are rarely found in healthy individuals or in patients with other connective tissue diseases [10]. When found in patients evaluated for Raynaud phenomenon, ACA have predictive value for those at risk for the future development of SSc [10•,14–16]. Conversely, in By contrast, patients with ACA do have an increased risk of pulmonary hypertension, though this is not due to abnormalities in coronary circulation [21,24•]. A recent study investigated whether determination of coronary flow reserve, as evaluated by transthoracic Doppler echocardiography, might represent a potential method of detecting early dysfunction of the cardiovascular system in patients with SSc without clinical signs or symptoms of cardiac impairment [24•]. Twenty-four out of 44 SSc patients showed reduced coronary flow reserve in comparison with a normal range of age- and sex-matched healthy subjects, especially those with diffuse cutaneous SSc, although no correlation was found with ACA. Patients who are ACA positive have a lower mortality than those with positive anti-Scl-70 autoantibodies or ANoA [20,25]. However, the relation of cancer to SSc is controversial. Although several studies have suggested an increased frequency of cancer in patients with SSc contributing to increased mortality, and two have correlated this risk with ACA, a more recent case-control study in a large cohort of patients with SSc was unable to replicate these findings [26–32,33•]. Table 1. Diagnostic and prognostic associations of autoantibodies Autoantibody Technique Predictive of Compared with ACA ACA ACA ACA ACA Anti-Scl-70 Anti-Scl-70 Anti-Scl-70 Anti-Scl-70 Anti-Scl-70 Anti-Scl-70 Anti-Scl-70 Anti-Scl-70 Anti-Scl-70 Anti-Scl-70 AFA Anti-RNAP PM-Scl IIF IIF IIF IIF IIF ID ID ID IB IB ELISA ELISA ID ID IP, ELISA, IB IB, IP IP, IB ID SSc SSc lcSSc Pulm. fibrosis SSc SSc SSc SSc SSc SSc SSc SSc dcSSc Pulm. fibrosis Pulm. fibrosis dcSSc dcSSc SSc/myositis overlap Healthy individuals Other CTDs Healthy individuals Healthy individuals RP Healthy individuals Other CTDs RP Healthy individuals Other CTDs Healthy individuals Other CTD Healthy individuals Healthy individuals Healthy individuals Healthy individuals Healthy individuals Healthy individuals Sensitivity % Specificity % Pos LR 44 31 44 12 24 20 26 28 41 40 43 43 37 45 43 12 38 50 99.9 97 93 71 90 100 99.5 98 99.4 99 100 90 82 81 83 97 94 98 327 12.5 6.1 0.41 2.3 > 25 52 10 68 40 > 55 4.3 2.0 2.3 2.5 4.0 6 31 ACA, anticentromere antibody; AFA, antifibrillarin antibody; RNAP, RNA polymerase; IIF, indirect immunofluorescence; ID, immunodiffusion; IB, immunoblotting; ELISA, enzyme-linked immunosorbent assay; IP, immunoprecipitation; SSc, systemic sclerosis; lcSSc, limited cutaneous systemic sclerosis; dcSSc, diffuse cutaneous systemic sclerosis; CTD, connective tissue disease; RP, Raynaud phenomenon; Pos LR, positive likelihood ratio. Published with permission [10]. Autoantibodies in systemic sclerosis Cepeda and Reveille 725 Once a patient is found to be ACA positive, there is no clinical utility in serial measurements. ACA-positive patients tend to remain positive over time, whether tested by indirect immunofluorescence or by immunoblotting [10••,34–36]. HLA-DRB1*01, HLA-DRB1*04, and HLA-DQB1*05 are associated with the presence of ACA, and it seems likely that the generation of ACA is influenced by the presence of both HLA-DRB1 and HLA-DQB1 alleles [2,12,37•]. Other MHC genes have also been implicated. A group from the United Kingdom has recently implicated tumor necrosis factor (TNF) genes, located in the MHC class III region, in the ACA response, specifically the TNF863A allele, the TNF-1031C allele, and with a TNF promoter haplotype, suggesting this to be the strongest non-HLA genetic marker so far described in SSc [37•]. Antitopoisomerase I antibodies Antitopoisomerase I (anti-Scl-70) antibodies have classically been determined by double immunodiffusion techniques against calf or rabbit thymus extract, including Ouchterlony and counterimmunoelectrophoresis. The Scl-70 antigen was found to be a basic, nonhistone chromosomal protein of 70,000 molecular weight found in rat liver, calf, or rabbit thymus, or in HEp-2 or lymphoid cells, and was subsequently found to represent topoisomerase I [10••]. Although initial ELISAs used topoisomerase I purified from calf thymus glands, more recent studies have used recombinant topo I fusion proteins as the substrate for the ELISAs [10••]. phenomenon can confer an increase in the future development of SSc [14,15]. As many as half of patients with SSc in whom pulmonary fibrosis develops will have Scl-70 autoantibodies, which in some studies have also been predicted more severe pulmonary disease (Table 1) [10,18–20]. A higher rate of decline in pulmonary function test results has been described in patients with anti-Scl-70, although this association is not universally seen [10••,39]. Despite a report in one study of Japanese patients with SSc, no convincing association has been established for anti-Scl-70 and scleroderma renal crisis [40]. Anti-Scl-70 antibodies carry an increased mortality rate, attributed to the higher rate of ventricular failure secondary to pulmonary disease [41,42]. As with ACA, a recent study did not show any effect of anti-Scl-70 antibodies on coronary flow reserve [24•]. As with ACA, an increased frequency of anti-Scl-70 in SSc patients in whom cancer develops has been reported previously [43]. However, a more recent case-control study in a large cohort of patients with SSc mentioned previously was unable to replicate these findings [33•]. The clinical utility of serial determinations of anti-Scl-70 antibodies is controversial. Some studies have shown patients initially seen to be Scl-70 positive and anti-Scl-70 negative remained so, whereas other studies revealed varying antibody levels over time [10••]. In a more recent study, Hu et al. [44•] examined the correlations between anti-Scl-70 antibody levels measured by ELISA with the degree and extent of skin thickening in a larger cohort of SSc patients and in SSc patients from whom multiple serum samples had been obtained. Serum levels of anti-Scl-70 antibodies correlated positively with disease severity in the skin (total Rodnan skin score) and with disease activity, on both cross-sectional and longitudinal analysis. When determined by immunodiffusion, anti-Scl-70 antibodies are found in 9 to 20% of patients with SSc and are highly disease specific (Table 1) [10••,12,13•,22•]. Anti-Scl-70 autoantibodies (by immunodiffusion) are virtually never seen in healthy control individuals, in nonaffected relatives of patients with SSc, or in patients with other connective tissue diseases or primary Raynaud phenomenon and are thus very useful in the diagnosis of SSc [10••]. The determination of anti-Scl-70 by ELISA is somewhat less specific for SSc, as illustrated in a more recent study where 25% of 128 patients with systemic lupus erythematosus (SLE) were found to be positive for anti-Scl-70 antibody by ELISA [38]. None of these SLE patients had an SSc overlap syndrome, and no relation was found with pulmonary fibrosis as is seen in scleroderma. Interestingly, the presence and levels of anti-Scl70 correlated with higher lupus activity scores, the presence of double-stranded DNA antibodies, and more pulmonary hypertension and renal involvement than in SLE patients without the antibody [38]. Antinucleolar antibodies The classic clinical association of anti-Scl-70 antibodies is with diffuse cutaneous involvement (Table 1) [10••,11,12,16]. As with ACA, the presence of anti-Scl70 antibodies in a patient initially evaluated for Raynaud Antinucleolar antibodies are defined by their characteristic appearance on indirect immunofluorescence and have been reported in 15 to 40% of patients with SSc [10••]. They are rarely detected in healthy control individuals [6,19,21]. The association of anti-Scl-70 antibodies and HLADRB1*1101/*1104 and DPB1*1301 is well described [2,12,45]. In a recent study of TNF polymorphisms in patients with SSc, anti-Scl-70 showed a positive association with the TNF-857T allele and a negative association with both TNF-1031C and TNF-863A [37•]. 726 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes Anti-PM-Scl The anti-PM-Scl autoantibody has been found in patients with myositis, scleroderma, and the polymyositisscleroderma overlap syndrome, which combines myositis, Raynaud phenomenon, arthritis, and interstitial lung disease [4]. In general, autoantibodies against the PMScl complex are found in approximately a quarter of patients with the polymyositis-scleroderma overlap syndrome, compared with only 2% of patients with scleroderma alone and 6% of patients with myositis (polymyositis or dermatomyositis) alone (Table 1) [4,46]. Conversely, between 43% and 88% of patients positive for anti-PM-Scl antibodies received diagnoses of myositis-scleroderma overlap syndrome [4,47]. The anti-PM-Scl autoantibody is directed against a multi-subunit nucleolar and nucleolar protein complex. The PM-Scl complex has been shown to be the equivalent of the yeast exosome, a complex consisting of at least 10 proteins, all displaying characteristics of exoribonucleases, which have been shown to be involved in the degradation and processing of many RNA species [48]. PM-Scl-100 and PM-Scl-75 contain the main autoantigenic epitopes of the human exosome. Recently, an N-terminally elongated PM-Scl-75 protein has been described [49]. Raijmakers et al. [50•] recently compared the autoantigenicity of the recently described Nterminally elongated PM-Scl-75 protein with that of PMScl-100 and the originally defined PM-Scl-75 polypeptide, and found that anti-PM-Scl-75 is more prevalent in patients with the polymyositis-scleroderma overlap syndrome than are anti-PM-Scl-100 autoantibodies, which until now were considered to be present most frequently. The clinical utility needs to be confirmed. Anti-Th/To Anti-Th/To antibodies are directed against components of the ribonuclease MRP and ribonuclease P complexes, more frequently Rp25 and hPop1. The Th40 autoantigen is identical to Rpp38 protein [51]. They are present in approximately 2 to 5% of patients with SSc and are associated with milder skin and systemic involvement (Table 1) [10••,42,51–54]. The marked exception is the more severe pulmonary fibrosis that has been reported in patients with anti-Th/To and resultant greater mortality [53–55]. In one study, anti-Th/To antibodies were increased in patients with xerophthalmia, esophageal dysmotility, and decreased DLCO [55]. The presence of anti-Th/To antibodies has been associated with HLADRB1*11 in some but not all studies [2,56]. Anti-Th/To has also been reported in localized scleroderma in one study, with its presence possibly indicating a mild form of cutaneous involvement [57]. [7–9,58,59]. Although they tend to coexist, their infrequency limits their utility in the diagnosis of SSc. AntiRNA polymerase II (RNAP II) autoantibodies have also been described in patients with SLE and overlap syndromes [60]. Anti-RNAP antibodies are associated with diffuse cutaneous involvement and SSc-related renal crisis, as well as with greater mortality [8,9,10••,42,61•]. A recent study analyzing sera from 115 Italian SSc patients found that anti-RNAP I and III were found to be less frequent than in Caucasian patients from the United Kingdom or the United States [61•]. However, only 2 patients from this series had scleroderma renal crisis, one of them having RNAP antibodies, which may explain the lower frequency of scleroderma renal crisis in this cohort. Previously, two reports examined HLA class II gene associations with anti-RNAP I/III antibodies in SSc patients [62,63]. Both studies failed to detect any statistically significant associations between the presence of anti-RNAP I/III antibodies and HLA-DRB1 or DQB1 alleles. More recently, Kuwana et al. [64•] found that the presence of anti-RNAP I/III antibodies was associated with HLA-DRB1*0405 and DQB1*0401 in Japanese SSc patients, and with HLA-DRB3*02 (formerly known as HLA-DR52b) in Caucasians. However, these associations were weak and inconsistent between these two ethnic groups. Antifibrillarin/anti-U3 RNP Anti-U3 RNP antibodies have been described in SSc patients as far back as 1988 [3]. More recently, it has been shown that the mammalian U3 small nuclear RNP is one member of a family of nucleolar small nuclear RNPs that are immunoprecipatable by antifibrillarin antibodies (AFA) [65]. AFA are present in fewer than 10% of patients with SSc and have also been described in patients with SLE, UCTD, and primary Raynaud phenomenon [19,55]. AFA are highly associated with diffuse cutaneous SSc (Table 1). AFA frequency is higher in patients of African descent with SSc than in Caucasian patients with SSc, and they have been associated with myositis, pulmonary hypertension, and renal disease [19,66,67]. Anti-U3-RNP antibodies have also been found in serum samples from patients with localized scleroderma [68]. However, no correlation between clinical and laboratory manifestations was found. In one study, U3-RNP was found to be associated with HLA-DQB1*0604 [67]. Anti-RNA polymerase I–III antibodies Anti-Ku antibodies Autoantibodies to RNA polymerase I and III (RNAP I and III, respectively) are highly specific for SSc, although they occur in only 20% of patients (Table 1) Originally, autoantibodies against the 80-kDa subunit protein of the human autoantigen Ku (p70/p80) (anti-Ku antibodies) were described in patients with scleroderma- Autoantibodies in systemic sclerosis Cepeda and Reveille 727 myositis overlap syndromes [69]. Subsequently, associations were established with pulmonary hypertension in this setting [70]. It is now known that anti-Ku antibodies can occur in a wide spectrum of rheumatic diseases and have little utility in clinical practice [71,72]. Antiphospholipid antibodies The frequency of antiphospholipid antibodies (aPL) in SSc is approximately 20 to 25% (ranging widely from 0 to 63%) [72,73]. There may be an increased frequency of pregnancy losses in SSc patients with aPL [74]. The presence of anticardiolipin antibodies (aCL) in patients with SSc has been associated with a history of thrombosis and pulmonary hypertension [75,76]. Some studies have correlated the presence of aCL with greater skin and systemic involvement, although this is not seen in others [73,77–79]. The role of aPL in pathogenesis and determining long-term outcomes in SSc presently is not clear because of limited and inconsistent data. Therefore, the clinical utility of determining aCL in patients with SSc has not yet been established. The frequency of anti-U1RNP antibodies in SSc is approximately 8% (ranging from 2 to 14%) [12,88,89]. High titers of anti-U1RNP antibodies are most often found in association with what was previously designated “mixed connective tissue disease” with a frequency of more than 90% [88,90–93]. Clinically, anti-U-1-RNP is associated with a benign course characterized by less cutaneous and renal involvement with a favorable response to corticosteroids [88,91]. Other clinical manifestations associated with U-1RNP include Raynaud phenomenon, puffy hands, sicca, pulmonary disease, esophageal disease, and cor pulmonale secondary to pulmonary hypertension [88–91,94]. U1-RNP antibodies have also been reported in serum samples from patients with localized scleroderma [95]. Anti-Ro antibodies Anti-Ro antibodies occur at a lower frequency in SSc (<35%) than in SLE or Sjögren syndrome [96]. Sjögren syndrome has been described in up to 20% of all patients with SSc, with about one third to one half of those with anti-Ro antibodies [97,98]. Only a few studies have been performed to examine the association between Raynaud phenomenon and aCL, with conflicting results [80,81–84]. A more recent study by Caccavo et al. [85] found that secondary Raynaud phenomenon is not positively associated with the presence of aPL in patients with SLE as well as a negative association between IgG aCL and RP. ANCA have been reported at a low incidence in SSc (∼3%) without any significantly associated clinical features [99]. Case reports of scleroderma with vasculitis are rarely seen, and routine screening of ANCA in SSc is not recommended. Another study of 48 patients investigated whether aPL were detected in patients with localized scleroderma [86]. In this study, patients with localized scleroderma exhibited aCL (46%) and (LAC) (24%), whereas B2GPI antibodies were not detected. 70% of patients with generalized morphea had aCL and LAC, suggesting that aCL and LAC may be the major autoantibodies in patients with generalized morphea. Despite the high prevalence of aCL and LAC, however, thrombosis was detected in only 1 patient with generalized morphea. Autoantibodies against endothelial cell antigen have also been described in patients with SSc. They have been found to be associated with alveolocapillary involvement, pulmonary hypertension, digital ulcers and ischemia, severe Raynaud phenomenon, and capillaroscopic abnormalities [100–102]. Anti-endothelial cell antibodies have also been found to be correlated with pulmonary fibrosis [103,104•]. Further studies are needed with this antibody to determine whether it is useful in clinical practice. Antibodies against extractable nuclear antigens Autoantibodies directed against fibrillin-1 protein, an extracellular matrix microfibrillar protein can occur in both localized scleroderma and in SSc [105]. In a prospective study analyzing serial serum samples, antifibrillin-1 autoantibody was present in 49% of those with limited scleroderma and 47% of those with mixed connective tissue disease 106]. Anti-Sm and anti-U1-RNP antibodies The presence of anti-Sm antibodies is considered to be highly specific for SLE, although they have been described as occurring uncommonly in patients with SSc [12,87,88]. By contrast, anti-U1-RNP antibodies are associated with a variety of connective tissue diseases, including SLE, SSc, polymyositis, and mixed connective tissue diseases [88]. When found in patients with SSc, anti-Sm antibodies are found most often with SLE overlap and carry a poorer prognosis and complications such as lupus nephritis, renal crisis, and pulmonary hypertension [87]. Less extensively studied autoantibodies A recent study showed anti-U1 RNA antibodies to be present in patients with SSc with anti-U1 RNP antibodies, mixed connective tissue disease, and SLE, but not in healthy control individuals [107]. These results indicate that anti-U1 RNA antibodies may be a serologic indicator for pulmonary fibrosis in SSc patients with anti-U1 RNP antibodies; however, clearly more studies are needed. 728 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes Fibrosing disorders The fibrosing disorders represent a diverse group of chronic, frequently progressive, systemic or localized diseases characterized by an over-accumulation of extracellular matrix components that interferes with function (Table 2). The cause of most fibrosing disorders is generally still a mystery. Despite the frequent occurrence of these diseases, the medical management of these disorders is inadequate, and our understanding of their pathogenesis is limited. The association with most of these fibrosing disorders and autoantibodies has for the most part been inconclusive. scleroderma and 100% of patients with generalized morphea [115]. Whether this is a marker for localized scleroderma remains to be determined. Graft-versus-host disease A variety of autoantibodies have been reported in patients with graft-versus-host disease, particularly when sclerodermatous skin changes are present, including discrepant reports of anti-Scl-70 (21% in one study but not in another), anti-PM-Scl, aCL, or ANCA, although their significance and clinical utility is unclear [116–120]. Eosinophilia myalgia syndrome Localized scleroderma Whereas anti-topoisomerase I antibody is almost exclusively detected in SSc, autoantibodies against topoisomerase-II-␣ have been detected in idiopathic pulmonary fibrosis, SSc, insulin-dependent diabetes mellitus, juvenile rheumatoid arthritis, and SLE [108–113]. A recent study suggests that anti-topo-II-␣ is a major autoantibody in localized scleroderma [114]. Further studies are needed with this autoantibody. As described above, aPL, anti-U1, U3-RNP, and antiTh/To autoantibodies have also been described in patients with localized scleroderma. Recently, a novel autoantibody to Cu/Zn superoxide dismutase was detected in 89% of patients with localized Table 2. Selected fibrosing disorders Skin and musculoskeletal system Systemic sclerosis Localized form of scleroderma Scleredema Scleromyxedema Keloids Dupuytren contracture Eosinophilic fasciitis Eosinophilia-myalgia syndrome Graft-versus-host disease Plantar fasciitis Nephrogenic fibrosing dermopathy Diabetic stiff-hand syndrome Radiation fibrosis Lungs Pulmonary fibrosis Chronic pleural reaction Cardiovascular system Constrictive pericarditis Intimal proliferation Gastrointestinal system Primary biliary cirrhosis Sclerosing cholangitis Collagenous colitis Genitourinary system Nephrosclerosis Peyronie disease Nephritis Other Reidel struma Retroperitoneal fibrosis Cancer Some past studies have suggested an association between eosinophilia myalgia syndrome and autoantibodies such as ANA and antiphospholipid antibodies [121,122]. However, the clinical significance of these antibodies remains unclear in this increasingly rare syndrome. Nephrogenic fibrosing dermopathy It is well known that patients with chronic renal failure and hemodialysis frequently have increased serum levels of antinuclear antibodies and circulating immune complexes. It is therefore difficult to assess the diagnostic validity of serologic autoimmune phenomena in patients undergoing hemodialysis. This is illustrated by the problem with aCL, which have been reported in nephrogenic fibrosing dermopathy [123]. However, elevated aCL may occur in approximately 30% of patients with chronic hemodialysis [124]. There have also been reports of nephrogenic fibrosing dermopathy associated with anti– double-stranded DNA antibodies [125]. Hence, there are few data to justify determining autoantibodies in patients with nephrogenic fibrosing dermopathy at this point. Conclusion Autoantibodies in SSc provide important and prognostic information (Table 3) and are useful in defining clinical subsets of the disease. ACA are very useful in the diagnosis of SSc, particularly in patients with limited cutaneous involvement, and rarely occur in those with pulmonary fibrosis. They are also associated with a better prognosis, whereas anti-Scl-70 antibodies, also useful in the diagnosis of SSc, predict diffuse skin involvement and pulmonary fibrosis and are associated with a poorer prognosis. SSc patients who express ANoA run the gamut of having mild disease (anti-PM-Scl) through limited skin but severe pulmonary involvement (antiTh/To), to intermediately severe disease (anti-fibrillarin) (anti-RNA polymerase I and III). Other autoantibodies (anti-RNP, anti-Ro) are less specific for SSc, although they do define clinical subsets (overlap syndromes and sicca complex, respectively). Still other autoantibodies have been reported whose clinical relevance is doubtful Autoantibodies in systemic sclerosis Cepeda and Reveille 729 Table 3. Autoantibodies in systemic sclerosis Prevalence in SSc HLA associations ACA 20 to 30% (highest frequency seen in Caucasians) HLA-DRB1*01, *04 HLA-DQB1*05 Anti-Scl-70 9 to 20% HLA-DRB1*1101, *1104 HLA-DPB1*1301 Anti-PM-Scl-70 24% of patients with PM/SSc overlap 2% in SSc alone Anti-Th/To Autoantibody Clinical and serologic associations Prognosis Other CREST lcSSc Lower frequency of pulm. fibrosis Increased risk of pulmonary hypertension dcSSc Increased frequency of pulm. fibrosis Lower mortality rate than anti-Scl-70 or ANoA ? Relation with cancer No need for serial measurements Increased mortality rate ? Relation with cancer Serial determinations controversial HLA-DQA1*0501 HLA-DRB1*0301 PM/SSc overlap 2 to 5% HLA-DRB1*11 Anti-RNAP 20% AFA < 10% ?HLA-DRB1*0405 ?HLA-DQB1*0401 ?HLA-DBR3*2 ?HLA-DQB1*0604 Milder skin and systemic involvement More severe pulmonary fibrosis dcSSc ?Renal crisis Benign and chronic course with favorable response to steroids Worse prognosis Anti-Ku antibodies Infrequent No known association aPL 20 to 25% (ranging widely from 0 to 63%) No known association Anti-Sm Rare Highly specific for SLE No known association Anti-U1RNP 8% HLA-DR2, DR4 Anti-Ro Infrequent ANCA Infrequent Antifibrillin-1 antibody Antiendothelial cell antibodies Infrequent HLA-DRB1*0301, DQA1*0501, DQB1*0201 No known association No known association No known association Infrequent dcSSc Myositis, pulmonary hypertension and renal disease Overlap syndrome with scleroderma features ? Thrombosis, pulmonary hypertension ? More skin involvement When found in patients with SSc, most often with SLE overlap, lupus nephritis, renal crisis, pulmonary hypertension MCTD, RP, puffy hands, myositis, pulmonary hypertension Sicca symptoms Increased mortality Poorer prognosis when found with SSc/SLE overlap More benign course Rare reports of SSC with vasculitis SSc, MCTD Limited scleroderma Alveolocapillary involvement pulmonary htn, severe RP, pulmonary fibrosis, digital ulcers ACA, anticentromere antibodies; SSc, systemic sclerosis; lcSSc, limited cutaneous systemic sclerosis; ANoA, antinucleolar antibodies; dcSSc, diffuse cutaneous systemic sclerosis; PM, polymyositis; RNAP, RNA-polymerase; AFA, antifibrillarin antibodies; aPL, antiphospholipid; MCTD, mixed connective tissue disease; ANCA, anti-neutrophil cytoplasmic antibodies. 730 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes (as a result of either their rare occurrence or the conflicting data), such as anti-Ku, antiphospholipid antibodies, anti-Sm, and ANCA. Newer autoantibodies systems, such as anti-endothelial cell antibodies, antifibrillin-1 autoantibody, and anti-U1 RNA antibodies, look interesting but need more study. Data on these autoantibodies and other fibrosing disorders have for the most part been inconclusive. Autoantibodies associated with SSc differ in their associated clinical manifestations, ethnic and genetic associations, pathophysiology, and frequencies. Although it remains controversial whether these autoantibodies have an actual role in pathogenesis, these serologic markers can be useful in the diagnosis and clinical management of SSc when backed by careful clinical judgment. References and recommended reading ethnic groups: a comparison of clinical, sociodemographic, serologic, and immunogenetic determinants. Semin Arthritis Rheum 2001, 30:332–346. Mayes MD, Lacey JV Jr, Beebe-Dimmer J, et al.: Prevalence, incidence, survival, and disease characteristics of systemic sclerosis in a large US population. Arthritis Rheum 2003, 48:2246–2255. The authors established baseline estimates of the prevalence, incidence, survival, and disease characteristics based on 706 verified cases of SSc consisting primarily of black and white adults. 13 • 14 Wollersheim H, Thien T, Hoet MH, et al.: The diagnostic value of several immunological tests for anti-nuclear antibody in predicting the development of connective tissue disease in patients presenting with Raynaud’s phenomenon. Eur J Clin Invest 1989, 19:535–541. 15 Kallenberg CG, Wouda AA, Hoet MH, et al.: Development of connective tissue disease in patients presenting with Raynaud’s phenomenon: a six year follow up with emphasis on the predictive value of antinuclear antibodies as detected by immunoblotting. Ann Rheum Dis 1988, 47:634–641. 16 Weiner ES, Earnshaw WC, Senecal JL, et al.: Clinical associations of anticentromere antibodies and antibodies to topoisomerase I: a study of 355 patients. Arthritis Rheum 1988, 31:378–385. 17 Cutolo M, Pizzorni C, Tuccio M, et al.: Nailfold videocapillaroscopic patterns and serum autoantibodies in systemic sclerosis. Rheumatology (Oxford) 2004, 43:719–726. 18 Steen VD, Powell DL, Medsger TA Jr, et al.: Clinical correlations and prognosis based on serum autoantibodies in patients with systemic sclerosis. Arthritis Rheum 1988, 31:196–203. 19 Ferri C, Valentini G, Cozzi F, et al.: Systemic sclerosis: demographic, clinical, and serologic features and survival in 1,012 Italian patients. Medicine (Baltimore) 2002, 81:139–153. 20 Hesselstrand R, Scheja A, Shen GQ, et al.: The association of antinuclear antibodies with organ involvement and survival in systemic sclerosis. Rheumatology (Oxford) 2003, 42:534–540. 21 Mitri GM, Lucas M, Fertig N, et al.: A comparison between anti-Th/To- and anticentromere antibody-positive systemic sclerosis patients with limited cutaneous involvement. Arthritis Rheum 2003, 48:203–209. Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest Feghali-Bostwick C, Medsger TA Jr, Wright TM: Analysis of systemic sclerosis in twins reveals low concordance for disease and high concordance for the presence of antinuclear antibodies. Arthritis Rheum 2003, 48:1956– 1963. This study examined concordance for SSc in monozygotic and dizygotic twins. Concordance for SSc was found to be similar in monozygotic and dizygotic twins, and overall concordance for SSc was low in the twins (4.7%). SSc-associated serum autoantibodies occurred exclusively in patients with SSc. The study showed that inheritance may play a role in the development of serum autoantibodies in the “healthy” twin sibling of a patient with SSc. 1 • 22 Allcock RJ, Forrest I, Corris PA, et al.: A study of the prevalence of systemic sclerosis in northeast England. Rheumatology (Oxford) 2004, 43:596–602. • This study estimated the prevalence and demographics of SSc, limited cutaneous SSc, diffuse cutaneous SSc, and anticentromere and anti-Scl-70 antibodies in northeast England. 2 Mayes MD, Reveille JD: Epidemiology, demographics, and genetics of systemic sclerosis. In: Systemic sclerosis. Edited by Clements PJ, Furst DE. Philadelphia: Lippincott Williams & Wilkins; 2003. 3 Reimer G, Steen VD, Penning CA, et al.: Correlates between autoantibodies to nucleolar antigens and clinical features in patients with systemic sclerosis (scleroderma). Arthritis Rheum 1988, 31:525–532. 23 4 Oddis CV, Okano Y, Rudert WA, et al.: Serum autoantibody to the nucleolar antigen PM-Scl. Clinical and immunogenetic associations. Arthritis Rheum 1992, 35:1211–1217. 5 Blaszcyk M, Jarzabek-Chlorzelska M, Jablonska S, et al.: Autoantibodies to nucleolar antigens in systemic scleroderma: clinical correlations. Br J Dermatol 1990, 123:421–430. 6 Reddy R, Tan EM, Henning D, et al.: Detection of a nucleolar 7-2 ribonucleoprotein and a cytoplasmic 8-2 ribonucleoprotein with autoantibodies from patients with scleroderma. J Biol Chem 1983, 258:1383–1386. 24 Sulli A, Ghio M, Bezante GP, et al.: Blunted coronary flow reserve in systemic sclerosis. Rheumatology (Oxford) 2004, 43:505–509. • The authors of this study investigated whether the noninvasive determination of coronary flow reserve, as evaluated by transthoracic Doppler echocardiography, might be a potential method to detect early dysfunction of cardiovascular system in 44 patients with SSc without clinical signs or symptoms of cardiac impairment. Coronary flow reserve was found to be lower in SSc patients, which may point to an indirect sign of heart involvement in SSc. 7 Reimer G, Rose KM, Scheer U, et al.: Autoantibody to RNA polymerase I in scleroderma sera. J Clin Invest 1987, 79:65–72. 8 Kuwana M, Kaburaki J, Mimori T, et al.: Autoantibody reactive with three classes of RNA polymerases in sera from patients with systemic sclerosis. J Clin Invest 1993, 91:1399–1404. 9 Okano Y, Steen VD, Medsger TA Jr, et al.: Autoantibody reactive with RNA polymerase III in systemic sclerosis. Ann Intern Med 1993, 119:1005–1013. Reveille JD, Solomon DH, American College of Rheumatology, ad hoc Committee on Immunologic Testing Guidelines.: Evidence-based guidelines for the use of immunologic tests: anticentromere, Scl-70, and nucleolar antibodies. Arthritis Rheum 2003, 49:399–412. This committee report analyzes the published literature on the clinical usefulness of SSc autoantibodies, the different technologies in ordering these autoantibodies, and the application of these autoantibodies in prognosis, specifically in predicting the extent of cutaneous and pulmonary involvement. 10 •• 11 12 McNeilage LJ, Youngchaiyud U, Whittingham S, et al.: Racial differences in antinuclear antibody patterns and clinical manifestations of scleroderma. Arthritis Rheum 1989, 32:54–60. Reveille JD, Fischbach M, McNearney T, et al.: Systemic sclerosis in 3 US Wigley FM, Wise RA, Miller R, et al.: Anticentromere antibody as a predictor of digital ischemic loss in patients with systemic sclerosis. Arthritis Rheum 1992, 35:688–693. 25 Scussel-Lonzetti L, Joyal F, Raynauld JP, et al.: Predicting mortality in systemic sclerosis: analysis of a cohort of 309 French Canadian patients with emphasis on features at diagnosis as predictive factors for survival. Medicine (Baltimore) 2002, 81:154–167. 26 Duncan SC, Winkelmann RK: Cancer and scleroderma. Arch Dermatol 1979, 115:950–955. 27 Peters-Golden M, Wise RA, Hochberg M, et al.: Incidence of lung cancer in systemic sclerosis. J Rheumatol 1985, 12:1136–1139. 28 Roumm AD, Medsger TA: Cancer and systemic sclerosis: an epidemiologic study. Arthritis Rheum 1985, 28:1336–1340. 29 Abu Shakra M, Guillemin F, Lee P, et al.: Cancer in systemic sclerosis. Arthritis Rheum 1993, 36:460–464. 30 Rosenthal AK, McLaughlin JK, Gridley G, et al.: Incidence of cancer among patients with systemic sclerosis. Cancer 1995, 76:910–914. 31 Higuchi M, Horiuchi T, Ishibashi N, et al.: Anticentromere antibody as a risk factor for cancer in patients with systemic sclerosis. Clin Rheumatol 2000, 19:123–126. 32 Kyndt X, Hebbar M, Queyrel V, et al.: Systemic scleroderma and cancer: search for predictive factors of cancer in 123 patients with scleroderma [in French]. Rev Med Interne 1997, 18:528–532. Autoantibodies in systemic sclerosis Cepeda and Reveille 731 Derk CT, Sakkas LI, Rasheed M, et al.: Autoantibodies in patients with systemic sclerosis and cancer: a case-control study. J Rheumatol 2003, 30:1994–1996. In this case-control study, the authors found no statistically significant difference in the frequency of ACA and anti-Scl-70 antibodies among patients with SSc with and without the diagnosis of cancer. This study conflicts with previous studies that certain autoantibodies may represent risk factors for the development of cancer in patients with SSc 33 • tibody reactivity in patients with systemic sclerosis and their relatives. J Rheumatol 1997, 24:477–484. 53 Kuwana M, Kimura K, Hirakata M, et al.: Differences in autoantibody response to Th/To between systemic sclerosis and other autoimmune diseases. Ann Rheum Dis 2002, 61:842–846. 54 Okano Y, Medsger TA Jr: Autoantibody to Th ribonucleoprotein (nucleolar 7-2 RNA protein particle) in patients with systemic sclerosis. Arthritis Rheum 1990, 33:1822–1828. 55 Van Eenennaam H, Vogelzangs JH, Bisschops L, et al.: Autoantibodies against small nucleolar ribonucleoprotein complexes and their clinical associations. Clin Exp Immunol 2002, 130:532–540. 34 Weiner ES, Hildebrandt S, Senecal JL, et al.: Prognostic significance of anticentromere antibodies and anti-topoisomerase I antibodies in Raynaud’s disease: a prospective study. Arthritis Rheum 1991, 34:68–77. 35 Tramposch HD, Smith CD, Senecal JL, et al.: A long-term longitudinal study of anticentromere antibodies. Arthritis Rheum 1984, 27:121–124. 56 36 Vasquez-Abad D, Russell CA, Cusick SM, et al.: Longitudinal study of anticentromere and antitopoisomerase-I isotypes. Clin Immunol Immunopathol 1995, 74:257–270. Falkner D, Wilson J, Medsger TA Jr, et al.: HLA and clinical associations in systemic sclerosis patients with anti-Th/To antibodies. Arthritis Rheum 1998, 41:74–80. 57 Yamane K, Ihn H, Kubo M, et al.: Antibodies to Th/To ribonucleoprotein in patients with localized scleroderma. Rheumatology (Oxford) 2001, 40:683– 686. 58 Chang M, Wang R, Yangco D, et al.: Analysis of autoantibodies against RNA polymerases using immunoaffinity-purifed RNA polymerase I, II, and III antigen in an enzyme-linked immunosorbent assay. Clin Immunol Immunopathol 1998, 89:71–78. 59 Bunn CC, Denton CP, Shi-Wen X, et al.: Anti-RNA polymerases and other autoantibody specificities in systemic sclerosis. Br J Rheumatol 1998, 37:15–20. 60 Satoh M, Kuwana M, Ogasawara T, et al.: Association of autoantibodies to topoisomerase I and the phosphorylated (IIO) form of RNA polymerase II in Japanese scleroderma patients. J Immunol 1994, 153:5838–5848. Sato H, Lagan AL, Alexopoulou C, et al.: The TNF-863A allele strongly associates with anticentromere antibody positivity in scleroderma. Arthritis Rheum 2004, 50:558–564. In this study of 214 SSc patients from the United Kingdom, the authors investigated TNF polymorphisms in scleroderma. Anti-Scl-70 showed a positive association with the TNF-857T allele and a negative association with both TNF-1031C and TNF-863A alleles. 37 • 38 Gussin HA, Ignat P, Varga J, et al.: Anti-topoisomerase I (anti-Scl-70) antibodies in patients with systemic lupus erythematosus. Arthritis Rheum 2001, 44:376–383. 39 Greidinger EL, Flaherty KT, White B, et al.: African-American race and antibodies to topoisomerase I are associated with increased severity of scleroderma lung disease. Chest 1998, 114:801–807. 40 Nishijima C, Sato S, Hasegawa M, et al.: Renal vascular damage in Japanese patients with systemic sclerosis. Rheumatol 2001, 40:406–409. 41 Murata I, Takenaka K, Shinohara S, et al.: Diversity of myocardial involvement in systemic sclerosis: an 8-year study of 95 Japanese patients. Am Heart J 1998, 135:960–969. 42 43 Jacobsen S, Ullman S, Shen GQ, et al.: Influence of clinical features, serum antinuclear antibodies, and lung function on survival of patients with systemic sclerosis. J Rheumatol 2001, 28:2454–2459. Rothfield N, Kurtzman S, Vasques-Abad D, et al.: Association of antitopoisomerase I with cancer. Arthritis Rheum 1992, 35:724. Hu PQ, Fertig N, Medsger TA Jr, et al.: Correlation of serum anti-DNA topoisomerase I antibody levels with disease severity and activity in systemic sclerosis. Arthritis Rheum 2003, 48:1363–1373. This article revisits the utility of the anti-Scl-70 ELISA in the care of patients with SSc. Contrary to some previous studies, the authors found that serum levels of anti-Scl-70 antibody (measured by ELISA) correlate positively with disease severity and disease activity in SSc. 44 • Bardoni A, Ossi P, Salvini R, et al.: Autoantibodies to RNA-polymerases in Italian patients with systemic sclerosis. Clin Exp Rheumatol 2003, 21:301– 306. In this study of Italian patients with SSc, the authors found anti-RNA-polymerase antibodies to be less frequent in Italian patients than in Caucasian patients from the United Kingdom or the United States. Also, anti-RNA-polymerase I and III antibodies were associated with diffuse cutaneous involvement and male gender. 61 • 62 Fanning GC, Welsh KI, Bunn C, et al.: HLA associations in three mutually exclusive autoantibody subgroups in UK systemic sclerosis patients. Br J Rheumatol 1998, 37:201–207. 63 Falkner D, Wilson J, Fertig N, et al.: Studies of HLA-DR and DQ alleles in systemic sclerosis patients with autoantibodies to RNA polymerases and U3RNP (fibrillarin). J Rheumatol 2000, 27:1196–1202. Kuwana M, Pandey JP, Silver RM, et al.: HLA class II alleles in systemic sclerosis patients with anti-RNA polymerase I/III antibody: associations with subunit reactivities. J Rheumatol 2003, 30:2392–2397. Previous studies have failed to detect any statistically significant associations between the presence of anti-RNAP I/III antibodies and HLA-DRB1 or DQB1 alleles. The authors of this study found that the presence of anti-RNAP I/III antibodies was associated with HLA-DRB1*0405 and DQB1*0401 in Japanese SSc patients, and with HLA-DRB3*02 in Caucasians. 64 • 65 Baserga SJ, Yang XD, Steitz JA: An intact Box C sequence in the U3 snRNA is required for binding of fibrillarin, the protein common to the major family of nucleolar snRNPs. EMBO J 1991, 10:2645–2651. 66 Arnett FC, Reveille JD, Goldstein R, et al.: Autoantibodies to fibrillarin in systemic sclerosis (scleroderma): an immunogenetic, serologic, and clinical analysis. Arthritis Rheum 1996, 39:1151–1156. 67 Tormey VJ, Bunn CC, Denton CP, et al.: Anti-fibrillarin antibodies in systemic sclerosis. Rheumatology (Oxford) 2001, 40:1157–1162. 68 Yamane K, Ihn H, Kubo M, et al.: Anti-U3 snRNP antibodies in localised scleroderma. Ann Rheum Dis 2001, 60:1157–1158. 69 Mimori T, Akizuki M, Yamagata H, et al.: Characterization of a high molecular weight acidic nuclear protein recognized by autoantibodies in sera from patients with polymyositis-scleroderma overlap. J Clin Invest 1981, 68:611– 620. 70 Isern RA, Yaneva M, Weiner E, et al.: Autoantibodies in patients with primary pulmonary hypertension: association with anti-Ku. Am J Med 1992, 93:307– 312. 71 Takeda Y, Dynan WS: Autoantibodies against DNA double-strand break repair proteins. Front Biosci 2001, 6:D1412–D1422. Raijmakers R, Renz M, Wiemann C, et al.: PM-Scl-75 is the main autoantigen in patients with the polymyositis/scleroderma overlap syndrome. Arthritis Rheum 2004, 50:565–569. When combining the long PM-Scl-75 isoform in addition to the PM-Scl-100 in ELISA, the authors noted that there is an increase in the number of patients in whom anti-PM-Scl autoantibodies can be detected. 72 Franceschini F, Cavazzana I, Generali D, et al.: Anti-Ku antibodies in connective tissue diseases: clinical and serological evaluation of 14 patients. J Rheumatol 2002, 29:1393–1397. 73 Picillo U, Migliaresi S, Marcialis MR, et al.: Clinical significance of anticardiolipin antibodies in patients with systemic sclerosis. Autoimmunity 1995, 20:1–7. 51 van Eenennaam H, Vogelzangs JHP, Lugtenberg D, et al.: Identity of the RNase MRP-and RNase P-associated Th/To autoantigen. Arthritis Rheum 2002, 46:3266–3272. 74 Black CM, Stevens WM: Pregnancy in patients with rheumatic diseases: scleroderma. Rheum Dis Clin North Am 1989, 15:193–212. 75 52 Harvey G, Black C, Maddison P, et al.: Characterization of antinucleolar an- Antonioli CM, Danieli E, Airo P, et al.: More on anticardiolipin and anti-beta2 glycoprotein I in systemic sclerosis. Ann Rheum Dis 2003, 62:589–590. 45 Gilchrist FC, Bunn C, Foley PJ, et al.: Class II HLA associations with autoantibodies in scleroderma: a highly significant role for HLA-DP. Genes Immun 2001, 2:76–81. 46 Brouwer R, Hengstman GJ, Vree Egberts W, et al.: Autoantibody profiles in the sera of European patients with myositis. Ann Rheum Dis 2001, 60:116– 123. 47 Marguerie C, Bunn CC, Copier J, et al.: The clinical and immunogenetic features of patients with autoantibodies to the nucleolar antigen PM-Scl. Medicine (Baltimore) 1992, 71:327–336. 48 Raijmakers R, Vree Egberts W, van Venrooij WJ, et al.: Protein-protein interactions between human exosome components support the assembly of Rnase PH-type subunits into a six-membered PNPase like ring. J Mol Biol 2002, 323:653–663. 49 Alderuccio F, Chan EK, Tan EM, et al.: Molecular characterization of an autoantigen of PM-Scl in the polymyositis/scleroderma overlap syndrome: a unique and complete human cDNA encoding an apparent 75-kD acidic protein of the nucleolar complex. J Exp Med 1991, 173:941–952. 50 • 732 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes 76 Ihn H, Sato S, Fujimoto M, et al.: Measurement of anticardiolipin antibodies by ELISA using beta 2-glycoprotein I (beta 2-GPI) in systemic sclerosis. Clin Exp Immunol 1996, 105:475–479. 77 Malia RG, Greaves M, Rowlands LM, et al.: Anticardiolipin antibodies in systemic sclerosis: immunological and clinical associations. Clin Exp Immunol 1988, 73:456–460. 78 Passaleva A, Massai G, Matucci-Cerinic M, et al.: Immunological abnormalities in a group of patients with limited cutaneous systemic sclerosis and prominent vascular disease. Autoimmunity 1990, 6:283–291. 79 Katayama I, Otoyama K, Kondo S, et al.: Clinical manifestations in anticardiolipin antibody-positive patients with progressive systemic sclerosis. J Am Acad Dermatol 1990, 23:198–201. 80 Merkel PA, Chang Y, Pierangeli SS, et al.: The prevalence and clinical associations of anticardiolipin antibodies in a large inception cohort of patients with connective tissue diseases. Am J Med 1996, 101:576–583. 81 Vayssairat M, Abuaf N, Baudot N, et al.: Abnormal IgG cardiolipin antibody titers in patients with Raynaud’s phenomenon and/or related disorders: prevalence and clinical significance. J Am Acad Dermatol 1998, 38:555– 558. 82 Naldi L, Locati F, Marchesi L, et al.: Cutaneous manifestations associated with antiphospholipid antibodies in patients with suspected primary antiphospholipid syndrome: a case-control study. Ann Rheum Dis 1993, 52:219–222. 83 Knapik-Kordecka M, Wysokinski WE: Clinical spectrum of Raynaud’s phenomenon in patients referred to vascular clinic. Cardiovasc Surg 2000, 8:457–462. 84 Bongard O, Bounameaux H, Miescher PA, et al.: Association of anticardiolipin antibodies and abnormal nailfold capillaroscopy in patients with systemic lupus erythematosus. Lupus 1995, 4:142–144. 85 Caccavo D, Del Porto F, Garzia P, et al.: Raynaud’s phenomenon and antiphospholipid antibodies in systemic lupus erythematosus: is there an association? Ann Rheum Dis 2003, 62:1003–1005. 86 Sato S, Fujimoto M, Hasegawa M, et al.: Antiphospholipid antibody in localised scleroderma. Ann Rheum Dis 2003, 62:771–774. 87 Kameda H, Kuwana M, Hama N, et al.: Coexistence of serum anti-DNA topoisomerase I and anti-Sm antibodies: report of 3 cases. J Rheumatol 1997, 24:400–403. 88 89 Sharp GC, Irvin WS, May CM: Association of antibodies to ribonucleoprotein and Sm antigens with mixed connective-tissue disease, systematic lupus erythematosus and other rheumatic diseases. N Engl J Med 1976, 295:1149– 1154. Ihn H, Yamane K, Yazawa N, et al.: Distribution and antigen specificity of anti-U1RNP antibodies in patients with systemic sclerosis. Clin Exp Immunol 1999, 117:383–387. 90 Lundberg I, Hedfors E: Clinical course of patients with anti-RNP antibodies: a prospective study of 32 patients. J Rheumatol 1991, 18:1511–1519. 91 Jablonska S, Blaszczyk M: Scleroderma overlap syndromes. Adv Exp Med Biol 1999, 455:85–92. 92 Frandsen PB, Kriegbaum NJ, Ullman S, et al.: Follow-up of 151 patients with high-titer U1RNP antibodies. Clin Rheumatol 1996, 15:254–260. 93 Rasmussen EK, Ullman S, Hoier-Madsen M: Clinical implications of ribonucleoprotein antibody. Arch Dermatol 1987, 123:601–605. 94 Kasukawa R: Mixed connective tissue disease. Intern Med 1999, 38:386– 393. 95 Yamane K, Ihn H, Kubo M, et al.: Anti-U1RNP antibodies in patients with localized scleroderma. Arch Dermatol Res 2001, 293:455–459. 96 Bell S, Krieg T, Meurer M, et al.: Antibodies to Ro/SSA detected by ELISA: correlation with clinical features in systemic scleroderma. Br J Dermatol 1989, 121:35–41. 97 Andonopoulos AP, Drosos AA, Skopouli FN, et al.: Sjogren’s syndrome in rheumatoid arthritis and progressive systemic sclerosis: a comparative study. Clin Exp Rheumatol 1989, 7:203–205. 98 Drosos AA, Andonopoulos AP, Costopoulos JS, et al.: Sjogren’s syndrome in progressive systemic sclerosis. J Rheumatol 1988, 15:965–968. 99 Ruffati A, Sinico RA, Radice A, et al.: Auto antibodies to proteinase 3 and myeloperoxidase in systemic sclerosis. J Rheumatol 2002, 29:918–923. 100 Pignone A, Scaletti C, Matucci-Cerinic M: Anti-endothelial cell antibodies in systemic sclerosis: significant association with vascular involvement and alveolo-capillary impairment. Clin Exp Rheumatol 1998, 16:527–532. 101 Negi VS, Tripathy NK, Misra R, et al.: Antiendothelial cell antibodies in sclero- derma correlate with severe digital ischemia and pulmonary arterial hypertension. J Rheumatol 1998, 25:462–466. 102 Salogin KV, LeTonqueze M, Saraux A, et al.: Antiendothelial cell antibodies: useful markers of systemic sclerosis. Am J Med 1997, 102:178–185. 103 Ihn H, Sato S, Fujimoto M, et al.: Characterization of autoantibodies to endothelial cells in systemic sclerosis (SSc): association with pulmonary fibrosis. Clin Exp Immunol 2000, 119:203–209. 104 Wusirika R, Ferri C, Marin M, et al.: The assessment of anti-endothelial cell antibodies in scleroderma-associated pulmonary fibrosis: a study of indirect • immunofluorescent and western blot analysis in 49 patients with scleroderma. Am J Clin Pathol 2003, 120:596–606. There are limited data on antiendothelial cell antibodies in scleroderma. However, this study is consistent with other studies showing a possible relation with antiendothelial cell antibodies and pulmonary fibrosis. 105 Tan FK, Arnett FC, Antohi S, et al.: Autoantibodies to the extracellular matrix microfibrillar protein, fibrillin-1, in patients with scleroderma and other connective tissue diseases. J Immunol 1999, 163:1066–1072. 106 Lundberg I, Antohi S, Takeuki K, et al.: Kinetics of anti-fibrillin-1 autoantibodies in MCTD and CREST syndrome. J Autoimmun 2000, 14:267–274. 107 Asano Y, Ihn H, Yamane K, et al.: The prevalence and clinical significance of anti-U1 RNA antibodies in patients with systemic sclerosis. J Invest Dermatol 2003, 120:204–210. 108 Grigolo B, Mazzetti I, Borzi RM, et al.: Mapping of topoisomerase II alpha epitopes recognized by autoantibodies in idiopathic pulmonary fibrosis. Clin Exp Immunol 1998, 114:339–346. 109 Meliconi R, Bestagno M, Sturani C, et al.: Autoantibodies to DNA topoisomerase II in cryptogenic fibrosing alveolitis and connective tissue disease. Clin Exp Immunol 1989, 76:184–189. 110 Grigolo B, Mazzetti I, Meliconi R, et al.: Anti-topoisomerase II alpha autoantibodies in systemic sclerosis-association with pulmonary hypertension and HLA-B35. Clin Exp Immunol 2000, 121:539–543. 111 Chang YH, Hwang J, Shang HF, et al.: Characterization of human DNA topoisomerase II as an autoantigen recognized by patients with IDDM. Diabetes 1996, 45:408–414. 112 Zuklys KL, Szer IS, Szer W, et al.: Autoantibodies to DNA topoisomerase II in juvenile rheumatoid arthritis. Clin Exp Immunol 1991, 84:245–249. 113 Hoffmann A, Heck MM, Bordwell BJ, et al.: Human autoantibody to topoisomerase II. Exp Cell Res 1989, 180:409–418. 114 Hayakawa I, Minoru H, Takehara K, et al.: Anti-DNA topoisomerase II alpha autoantibodies in localized scleroderma. Arthritis Rheum 2004, 50:227–232. 115 Nagai M, Hasegawa M, Takehara K, Sato S: Novel autoantibody to Cu/Zn superoxide dismutase in patients with localized scleroderma. J Invest Dermatol 2004, 122:594–601. 116 Bell SA, Faust H, Mittermuller J, et al.: Specificity of antinuclear antibodies in scleroderma-like chronic graft-versus-host disease: clinical correlation and histocompatibility locus antigen association. Br J Dermatol 1996, 134:848– 854. 117 Chosidow O, Bagot M, Vernant JP, et al.: Sclerodermatous chronic graftversus-host disease: analysis of seven cases. J Am Acad Dermatol 1992, 26:49–55. 118 Quarantara S, Shulman H, Ahmed A, et al.: Autoantibodies in human chronic graft-versus-host disease after hematopoietic cell transplantation. Clin Immunol 1999, 91:106–116. 119 Chan EY, Lawton JW, Lie AK, et al.: Autoantibody formation after allogeneic bone marrow transplantation: correlation with the reconstitution of CD5+ B cells and occurrence of graft-versus-host disease. Pathology 1997, 29:184– 188. 120 Martin SJ, Audrain MA, Oksman F, et al.: Antineutrophil cytoplasmic antibodies (ANCA) in chronic graft-versus-host disease after allogeneic bone marrow transplantation. Bone Marrow Transplant 1997, 20:45–48. 121 Kaufman LD, Varga J, Gomez-Reino JJ, et al.: Autoantibodies in sera from patients with L-tryptophan-associated eosinophilia-myalgia syndrome: demonstration of unique antigen-antibody specificities. Clin Immunol Immunopathol 1995, 76:115–119. 122 Carreira PE, Montalvo MG, Kaufman LD, et al.: Antiphospholipid antibodies in patients with eosinophilia myalgia and toxic oil syndrome. J Rheumatol 1997, 24:69–72. 123 Mackay-Wiggan JM, Cohen DJ, Hardy MA, et al.: Nephrogenic fibrosing dermopathy (scleromyxedema-like illness of renal disease). J Am Acad Dermatol 2003, 48:55–60. 124 Valeri A, Joseph R, Radhakrishnan J, et al.: A large prospective survey of anti-cardiolipin antibodies in chronic hemodialysis patients. Clin Nephrol 1999, 51:116–121. 125 Gambichler T, Paech V, Kreuter A, et al.: Nephrogenic fibrosing dermopathy. Clin Exp Dermatol 2004, 29:258–260. Cellular origins of fibroblasts: possible implications for organ fibrosis in systemic sclerosis Arnold E. Postlethwaitea,b, Hidenobu Shigemitsub,c and Siva Kanangata,c Purpose of review There is an intense interest in the potential of circulating blood cells and epithelium-related nonfibroblast cells to change into matrix synthesizing fibroblasts and myofibroblasts. These sources of fibroblasts may have importance in systemic sclerosis (scleroderma). Recent findings Epithelial cells from different sources can transition into fibroblasts and myofibroblasts in response to transforming growth factor  and other growth factors/cytokines. This is called epithelial-mesenchymal transition (EMT). EMT has been repeatedly demonstrated to occur in several models of renal fibrosis including lupus prone mice. Quite unexpectedly, bone morphogenic protein 7 prevents EMT and protects lupus mice and other renal fibrosis models from developing fibrosis in the kidneys. Human peripheral blood mononuclear cells under different conditions of culture give rise to several different types of fibroblast-like cells. In SSc, it has been observed that the sera have low levels of serum amyloid protein. Serum amyloid protein has been found to inhibit the generation of fibrocytes from CD14+ precursors. The implications of these potential sources of fibroblasts and myofibroblasts in systemic sclerosis and related rheumatic diseases are discussed. Summary Fibroblasts and myofibroblasts in skin and internal organs of patients with systemic sclerosis and related diseases may possibly arise not only from the resident fibroblast population but from epithelial cells, pericytes, monocytes, and other progenitors from the circulating pool of hematopoietic cells and stem cells. These alternative sources of fibroblasts would best be treated by specifically targeting the transition or transdifferentiation process by which cells change into fibroblasts. Keywords epithelial-mesenchymal transition, transition, transdifferentiation, monocyte, scleroderma Curr Opin Rheumatol 16:733–738. © 2004 Lippincott Williams & Wilkins. a Division of Connective Tissue Diseases, University of Tennessee Health Science Center, Memphis, Tennessee, USA; bDivision of Pulmonary Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; and cDepartment of Veterans Affairs Medical Center, Memphis, Tennessee, USA Correspondence to Arnold E. Postlethwaite, MD, Division Connective Tissue Diseases, University of Tennessee Health Science Center, 956 Court Avenue, Room G326, Memphis, TN 38163, USA Tel: 901 448 4979; fax: 901 448 7265; e-mail: [email protected] This work was supported in part by a VA Merit Review Grant, an NIAMS Scleroderma SCOR USPHS AR44890 grant, and a NIAMS/NIAID grant NO1 AR92242. Current Opinion in Rheumatology 2004, 16:733–738 Abbreviations EMT FLCs MOMPs PBMCs SSc SLE TGF- epithelial-mesenchymal transition fibroblast-like cells monocyte-derived mesenchymal progenitors peripheral blood mononuclear cells systemic sclerosis systemic lupus erythematosus transforming growth factor  © 2004 Lippincott Williams & Wilkins 1040–8711 Introduction Fibroblasts are mesenchymally derived spindle-shaped cells that synthesize the major interstitial fibrillar collagens that provide structure to organs and tissues of the body [1]. They are critical for wound repair and maintenance of the connective tissue matrix. Fibroblasts from different anatomic locations in human adults and fetuses differ remarkably in the genes that they express [2–5]. It is suggested from microarray analyses that fibroblasts from different anatomic sites should be considered distinct differentiated cell types [2]. Remarkably, HOX genes (a family of highly conserved transcription factors), which function during embryogenesis to determine positional orientation of differentiating cells, were found in adult fibroblasts to retain main characteristics of HOX gene expression patterns used during embryogenesis, suggesting that there is imprinting of fibroblasts [2]. Myofibroblasts develop from fibroblasts in response to stimulation of transforming growth factor  (TGF-) and other growth factors and cytokines [6]. Myofibroblasts are highly contractile and are essential for contracting wounds. They express ␣-smooth muscle actin, which plays a major role in effecting these contractile properties [6]. Increased numbers of fibroblasts and myofibroblasts with excessive matrix disposition in skin and involved internal organs are a hallmark of the prototypic fibrosing disease systemic sclerosis (scleroderma, SSc) and fibrotic complications of other rheumatic diseases (e.g., pulmonary fibrosis associated with rheumatoid arthritis, systemic lupus erythematous [SLE] and polymyositis and end-stage renal disease associated with SLE). Although great advances have been made in characterizing the 733 734 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes plethora of growth factors, cytokines, and other inflammatory mediators that regulate fibroblast growth and function [1], until recently, little attention has been paid to the cellular origins of fibroblasts and myofibroblasts that populate fibrotic tissues. It is now apparent that fibroblasts arise not only from proliferation of resident fibroblasts but can metamorphose from different cell types (e.g., circulating fibrocytes, CD14+ monocytes, pericytes, epithelial cells, and hepatic stellate cells) as fibrosis develops. In this article, we briefly review this topic. TGF-1, TGF-2, basic fibroblast growth factor, platelet-derived growth factor AB-AA-BB, and epidermal growth factor, resulting in fibrosis or scar formation [1]. Fibroblasts are quite diverse in their phenotypic, morphologic and synthetic response to cytokines, growth factors, and matrix molecules [10–12,13••,14,15], and, therefore, the cytokines and growth factors mediating fibrosis may not be the same in different locations in the skin and other organs. Could such fibroblast heterogeneity explain differences in locations of fibrosis in subtypes of SSc? Origins of fibroblasts The circulating pool Tissue-specific fibroblast precursor cells Historically, Metchnikoff [16] in 1892 reported that blood mononuclear cells transformed into connective tissue cells during cicatrization in the tail fin of tadpoles. Later, Maximov [17] in 1928 confirmed the findings of Metchnikoff. There was criticism from skeptics that the results of Metchnikoff and Maximov were the result of contaminating fibroblasts entering the blood sample and that, in culture, these fibroblasts would expand. This skepticism prevailed until the issue was revisited by Labat et al. [18] and Bucala et al. [19] in the early 1990s. The most extensively studied fibroblast precursors are epithelial cells. This topic was recently reviewed in detail by Kalluri and Neilson [7••]. In studies using radiation bone marrow chimeras, transgenic receptor mice and the unilateral ureter obstruction model of renal fibrosis, it was observed that 15% of fibroblasts in the fibrotic regions of the unilateral ureter obstruction kidney were derived from bone marrow, and 36% were derived from tubular epithelium by a process termed epithelialmesenchymal transition (EMT) [7••]. EMT occurs in developing vertebrate embryos and is a mechanism by which epithelial cells are released to migrate to various parts of the embryo where they undergo differentiation into other structures [8]. EMT is initiated by cytokines (e.g., TGF-, fibroblast growth factor-2, epidermal growth factor, insulinlike growth factor-II) and proteases that degrade basement membranes beneath epithelia [9–12]. The epithelial cells lose their polarity, tight junctions, adherence junctions, desmosomes, and cytokeratin intermediate filaments and then acquire a phenotype suited for migration that includes rearrangement of F-actin stress fibers and expression of fibropodia and lamellipodia [7••]. They then acquire fibroblast characteristics and express fibroblast-specific protein-1) [7••]. EMT in the kidney is inhibited by bone morphogenic protein-7, and EMT likely contributes to renal fibrosis in murine models of SLE [13••]. EMT may also play a role in pulmonary fibrosis in circumstances in which pulmonary epithelial cells might transition into fibroblasts/myofibroblasts. An additional possible tissuespecific fibroblast progenitor includes hepatic stellate cells, which are related to their more ubiquitous perivascular relative, the pericyte, and which are thought to transdifferentiate into myofibroblasts and contribute to the fibrogenesis of cirrhosis [14]. Leftovers from embryonic development The prevailing view for decades has been that fibroblasts populating normal tissues are derived from mesenchyme left over when organs were formed during fetal development. During inflammation and tissue injury, these resident fibroblasts proliferate and upregulate matrix synthesis in response to cytokine and growth factors such as Neo-fibroblasts Labat et al. [18] reported that blood monocytes from patients’ with osteomyelosclerosis and Engelmann disease in culture spontaneously transformed into fibroblast-like cells (FLCs) he termed neo-fibroblasts. These neo-fibroblasts arose from HLA-DR+ monocytes cultured from the blood of patients with cystic fibrosis [20]. These neo-fibroblasts were found to be pluripotent and secreted not only collagen type I but also uromodulin, amyloid- peptide, ␣-fetoprotein, and carcinoembryonic antigen [21], reminiscent of the pluripotent stem cells reported by Zhao et al. [22•] discussed below. From normal donors, neo-fibroblast generation was enhanced by soluble factors from T cells but was short-lived, surviving in culture only 17 days, and reverted to a macrophage phenotype after contact with T cells [18,20]. Fibrocytes Bucala et al. [19] have described a FLC that they call the “fibrocyte.” Fibrocytes are present in small numbers in human and murine peripheral blood and are optimally isolated by culturing peripheral blood mononuclear cells (PBMCs) in medium with a low serum concentration on tissue culture surfaces coated with fibronectin [17]. They express collagen type I, CD11b, CD13, CD34, CD45 RO, major histocompatibility complex class II and CD86 but are negative for ␣-smooth muscle actin [19]. Fibrocytes are negative for monocyte markers (CD14 and CD16) as well as being negative for dendritic cell markers (CD25, CD10, CD38) and pan-B cell antigen CD19 [19]. Abe et al. [23] found that CD14+ human blood monocytes in the presence of T cells gives rise to fibrocytes and that TGF-1 can induce fibrocytes to assume Cellular origins of fibroblasts Postlethwaite et al. 735 a myofibroblast phenotype expressing ␣-smooth muscle actin and contracting collagen gels. Studies using transwell cultures showed that T cells had to be in physical contact with the CD14+ monocytes for fibrocytes to develop [23]. Fibrocytes have some functional differences from normal dermal fibroblasts. For example, they chemotax to interleukin-1 but not to tumor necrosis factor ␣, TGF-1, or platelet-derived growth factor, recognized chemoattractants for dermal fibroblasts [23–26]. The response of fibrocytes to cytokines with regards to collagen synthesis also differs from that of fibroblasts in that interleukin-1 inhibits, whereas tumor necrosis factor ␣ stimulates, collagen synthesis, opposite from dermal fibroblast responses to these cytokines [24,27]. Pluripotent stem cells There is increasing interest in blood mononuclear nonT, non-B, and non-NK cells as progenitors of not only fibroblasts but of different cell types. Zhao et al. [22•] claim in a recent article that normal CD14+ enriched human blood mononuclear cells grown in eight-well Lab Tech chamber slides coated with collagen and stimulated repeatedly with monocyte colony-stimulating factor and leukemia inhibitory factor assume a spindleshaped morphology after 7 days in culture. These cells stained positive for CD14, CD45, CD34, and CI and were called pluripotent stem cells by these authors [22•]. These pluripotent stem cells were not further characterized as to the monocyte subset from which they arise or what cellular markers or functional properties they might share with fibroblasts and/or myofibroblasts (e.g., production of collagen type I, hyaluronic acid, prostaglandin E2, matrix metalloproteinase-1, tissue inhibitor of metalloproteinase, proliferation or chemotaxis in response to various cytokines/growth factors, contraction of collagen gels). Interestingly, these pluripotent stem cells were able to be differentiated into CD3+/CD8+ T cells after exposure to interleukin-2 and could differentiate into epithelial cells when cultured with epidermal growth factor, neuronal cells when cultured with nerve growth factor, endothelial cells when cultured with vascular endothelial growth factor, and hepatocytes when cultured with hepatocyte growth factor, and, quite remarkably, could be converted to macrophages on exposure in culture to lipopolysaccharide [22•]. Monocyte-derived mesenchymal progenitors A related study by Kuwana et al. [28•] described a spindle-shaped cell that was CD14+/CD45+/CD34+/CI+ and derived from CD14+ monocytes. They called these cells monocyte-derived mesenchymal progenitors (MOMPs). In their hands, MOMPs arose in culture only when PBMCs were cultured on fibronectin-coated surfaces in the presence of low-glucose Dulbecco modified Eagle medium supplemented with 10% fetal calf serum. For generation of MOMPs, there were absolute requisites for growing PBMCs on a fibronectin-coated surface and exposure to a soluble factor(s) from CD14− blood cells [28•]. By electron microscopic examination, MOMPs had structural components representing a mixture of features of phagocytes, mesenchymal, and endothelial cells [28•]. MOMPs stopped growing after five subpassages or 4 weeks in culture [26]. MOMPs could be made to differentiate along mesenchymal cell lineages into osteoblasts, myocytes, chondrocytes, and adipocytes by appropriate additives to growth medium [28•]. Analysis by flow cytometry or immunohistochemistry revealed that MOMPs also express additional monocyte markers (CD13, CD11b/Mac-1, CD11c, CD64), HLA class I and HLA-DR, costimulatory molecules (CD40, CD86), adhesion molecules (CD29, CD44, and CD54), stem cell markers (CD105/SH2), endothelial cell markers (CD31, CD144, Flt-1, Ac-LDL), and mesenchymal cell markers (type III collagen, fibronectin, vimentin) [28•]. MOMPs were not further characterized as to the functions that they might share with fibroblasts/myofibroblasts. Bone marrow has recently been shown to be a source of fibroblasts in the bleomycin pulmonary fibrosis model in vivo [29••]. Fibroblast progenitors: scleroderma and related diseases Possible participation in fibrosis associated with scleroderma and related rheumatic diseases of fibroblasts derived from sources other than those in the resident pool has received little attention to date, but investigations are ongoing to explore this issue. In this regard, Pilling et al. [30••] observed that serum amyloid protein inhibits outgrowth of fibrocytes from PBMCs and that sera from patients with SSc have reduced levels of serum amyloid protein [29••]. In abstracts that we have submitted for presentation at the 2004 meeting of the American College of Rheumatology, we report that we have observed that PBMCs from patients with SSc generate large numbers of FLCs, which are derived from CD14+ monocytes when cultured with type I collagen compared with controls. Furthermore, there is an inverse relationship between the degree to which FLCs grow from PBMCs of patients with diffuse SSc to the DLCO, suggesting that increased outgrowth of FLCs from SSc PMBCs might be a marker for pulmonary fibrosis in diffuse SSc. The FLCs that we observe from SSc PMBCs cultured with collagen type I appear to be different from classic fibrocytes in that they are CD14+/CD34−/CI+. A list of some fibroblast and mesenchymal cell progenitors is given in Table 1. In Figure 1, we present a diagram showing events that could lead to transdifferentiation of circulating monocytes into FLCs in SSc. If circulating monocytes are eventually shown to play a significant role in the fibrogenesis associated with SSc, then treatment of SSc could 736 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes Table 1. Comparison of fibroblast-like cells from systemic sclerosis with other known possible sources of circulating fibroblast and mesenchymal progenitors Cell markers FLC from SSc Fibrocyte MOMP Type I collagen CD14 CD45 CD11b CD34 Culture conditions + + + + − Grows in serum and plastic-coated culture plates; PBMC-derived factors for growth + − + + + Requires serum-free medium for optimal growth Survival Months Published data for weeks + + + + + Requires culture plates to be coated with fibronectin and CD14− PBMC soluble factors Weeks Mesenchymal stem cell Monocytederived PSC + − − − − Grows in plastic-coated plates Unknown + + Unknown + Requires MCSF and LIF Months Published data for weeks FLC, fibroblast-like cell; SSc, systemic scleroderma; MOMP, monocyte-derived mesenchymal progenitor; PSC, pluripotent stem cell; PBMC, peripheral blood mononuclear cell; MCSF, monocyte colony-stimulating factor; LIF, leukemia inhibitory factor. be directed at several potential sites in the process. These would include (1) the endothelial cell activation and upregulation of adhesion, which allow circulating T cells and monocytes to attach and migrate through small blood vessels into perivascular sites; (2) the T cell, which has to produce cytokines for transdifferentiation of SSc monocytes to occur; and (3) the CD14+ monocyte precursors of the FLCs. Several of the ongoing and planned clinical trials in SSc funded by the National Institutes of Health might intervene in the process. The oral tolerance trial of type I bovine collagen could tolerize the T cell to type I collagen and other T-cell antigens in patients with SSc, essentially shutting down cytokine production by T cells. The autologous stem cell transplant trial in SSc, soon to begin, might have a similar effect on T cells but potentially could convert endothelial cell abnormalities and monocyte abnormalities in the disease by replacing these with naïve endothelial cells and monocytes of a younger age. The pannus of the rheumatoid joint might develop in part from transdifferentiation of blood monocytes into synovial fibroblasts. The particular cytokine milieu in the rheumatoid arthritis synovium might favor transdifferentiation of blood monocytes into a synovial fibroblast with marked propensity to synthesize matrix metalloproteinase that degrade cartilage ligands and tendons. Immune reaction in the lungs of patients with rheumatoid arthritis, SLE, polymyositis, and SSc might set the Figure 1. Endothelium is damaged in various parts of the body in patients with systemic sclerosis, resulting in upregulation of vascular adhesion molecules that bind receptors in circulating T cells and monocytes These cells then migrate through vessel walls into the interstitium where they come in contact with type I collagen (CI) and other antigens triggers release of cytokines and growth factors from the T cells and monocytes. Monocytes respond to transdifferentiation into fibroblasts and myofibroblasts, which synthesize new matrix. Cellular origins of fibroblasts Postlethwaite et al. 737 stage for trafficking of circulating fibroblast progenitors such as CD14 monocytes and fibrocytes to the lung parenchyma where the cytokine environment would favor transdifferentiation of the progenitor into high matrix producing fibroblasts/myofibroblasts. The glomerular damage in SLE might facilitate trafficking of fibroblast progenitors into the glomerular structure, and tubular epithelial cells by EMT could participate in renal fibrosis to lead to an end-stage fibrotic kidney. Renal failure in SSc might be owing in part to EMT of tubular epithelial cells in response to injury and cytokine and growth factor signals generated in the SSc kidney. The possibility that fibroblasts might originate from circulating progenitors and from EMT in patients with SSc and related rheumatic diseases characterized by lung and renal fibrosis is intriguing and certainly deserves further investigation. A must read for the latest review on EMT with a heavy emphasis on the relationship to developmental biology and renal fibrosis. There are extensive references. 8 Hay ED: An overview of epithelio-mesenchymal transformations. Acta Anat 1995, 154:8–20. 9 Fan JM, Ng YY, Hill PA, et al.: Transforming growth factor-beta regulates tubular epithelial-myofibroblast transdifferentiation in vitro. Kidney Int 1999, 56:1455–1467. 10 Okada H, Dannoff TM, Kalluri R, et al.: The early role of FSP1 in epithelialmesenchymal transformation. Am J Physiol 1997, 273:563–574. 11 Moralie OG, et al.: IGF-II induces rapid beta-catenin relocation to the nucleus during epithelium to mesenchyme transition. Oncogene 2001, 20:4942– 4950. 12 Strutz F, et al.: Role of basic fibroblast growth factor-2 in epithelialmesenchymal transformation. Kidney Int 2002, 61:1714–1728. Zeisberg M, Bottiglio C, Kumar N, et al.: Bone morphogenic protein-7 inhibits progression of chronic renal fibrosis associated with two genetic mouse models. Am J Physiol Renal Physiol 2003, 285:F1060–F1067. Very interesting article that shows that bone morphogenic protein-7 protects mice with deficiency of ␣3-chain of type IV collagen and MLR/MpJlpr/lpr lupus mice from developing chronic renal fibrosis. 13 •• 14 Cassiman D, Libbrecht L, Desmet V, et al.: Hepatic stellate cell/myofibroblast subpopulation in fibrotic human and rat livers. J Hepatol 2002, 36:200–209. 15 Goldring SR, Stephenson ML, Downie E, et al.: Heterogeneity in hormone responses and patterns of collagen synthesis in cloned dermal fibroblasts. J Clin Invest 1990, 85:798–803. 16 Metchnikoff E: Lecons sur la pathologie comparee de l’inflammation. (Données à I’Institut Pasteur en avril et mai 1881) In: Bibliothèque des Annales de I’Institut Pasteur. Masson, Paris, 111, 1882 17 Maximov A: Culture of blood leucocytes. From lymphocytes and monocytes to connective tissue. Arch Exp Zellforsch 1928, 5:12. 18 Labat ML, Bringuier AF, Séébold-Choqueux C, et al.: Monocytic origin of fibroblasts: spontaneous transformation of blood monocytes into neofibroblastic structures in osteomyelosclerosis and Engelmann’s disease. Biomed Pharmacother 1991, 45:289. 19 Bucala R, Spiegel LA, Chesney J, et al.: Circulating fibrocytes define a new leukocyte subpopulation that mediates tissue repair. Mol Med 1994, 1:71. 20 Labat ML, Bringuier AF, Séébold-Choquex C, et al.: Cystic fibrosis: production of high levels of uromodulin-like protein by HLA-DR blood monocytes differentiating towards a fibroblastic phenotype. Biomed Pharmacother 1991, 45:387. 21 Bringuier AF, Séébold-Choqueux C, Moricard Y, et al.: T-lymphocyte control of HLA-DR blood monocyte differentiation into neo-fibroblasts. Further evidence of pluripotential secreting functions of HLA-DR monocytes, involving not only collagen but also uromodulin, amyloid- peptide, ␣-fetoprotein and carcinoembryonic antigen. Biomed Pharmacother 1992, 46:91–108. Conclusion Little, if any, progress has been made to date on halting or reversing the fibrosis characteristic of SSc and related rheumatic diseases. Perhaps we have been focusing on the wrong targets. The observations that circulating fibroblast and mesenchymal progenitors play roles in some animal models of fibrosis and that epithelial cells by EMT can assume a fibroblast/myofibroblast phenotype are ample reason to stimulate further study into the role of these alternative sources of fibroblasts in SSc and related diseases. Acknowledgments The authors thank Ginny Geer for the typing of the manuscript and constructing the figure. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest 1 Postlethwaite AE, Kang AH: Fibroblasts and matrix proteins. In: Inflammation: Basic Principles and Clinical Correlation. Edited by Gallin JI, Goldstein IM, Snyderman R. New York: Raven Press; 1992:747–773. 2 Chiang HY, Chi JT, Dudoit S, et al.: Diversity, topographic differentiation and positional memory in human fibroblasts. Proc Natl Acad Sci U S A 2002, 99:12877–12882. 3 Müller GA, Rodemann HP: Characterization of human renal fibroblasts in health and disease. I. Immunophenotyping of cultured tubular epithelial cells and fibroblasts derived from kidneys with histologically proven interstitial fibrosis. Am J Kidney Dis 1991, 17:680–683. 4 Garrett DM, Conrad GW: Fibroblast-like cells from embryonic chick cornea, heat, and skin are antigenically distinct. Dev Biol 1979, 70:50–70. 5 Dugina V, Alexandrova A, Chaponnier C, et al.: Rat fibroblasts cultured from various organs exhibit differences in alpha-smooth muscle actin expression, cytoskeletal pattern, and adhesive structure organization. Exp Cell Res 1998, 238:481–490. 6 Serini G, Gabbiani G: Mechanisms of myofibroblast activity and phenotypic modulation. Exp Cell Res 1999, 250:273–283. 7 •• Kalluri R, Neilson EG: Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 2003, 112:1776–1784. Zhao Y, Glesne D, Huberman E: A human peripheral blood monocytesderived subset acts as pluripotent stem cells. Proc Natl Acad Sci U S A 2003, 100:2426–2431. This paper demonstrates the tremendous plasticity of the human CD14+ monocyte with its ability to differentiate into a variety of mesenchyme-related cells. 22 • 23 Abe R, Donnelly SC, Peng T, et al.: Peripheral blood fibrocytes: differentiation pathway and migration to wound sites. J Immunol 2001, 166:7556–7562. 24 Postlethwaite AE, Raghow R, Stricklin GP, et al.: Modulation of fibroblast functions by interleukin-1: increased steady state accumulation of type I procollagen mRNAs and stimulation of other functions but not chemotaxis by human recombinant interleukin 1␣ and . J Cell Biol 1988, 106:311–318. 25 Postlethwaite AE, Keski-Oja J, Moses HL, et al.: Stimulation of the chemotactic migration of human fibroblasts by transforming growth factor . J Exp Med 1987, 165:251–256. 26 Senior RM, Huang JS, Griffin GL, et al.: Dissociation of the chemotactic and mitogenic activities of PDGF by human neutrophil elastase. J Cell Biol 1985, 100:351–356. 27 Chesney J, Metz C, Stavitsky AB, et al.: Regulated production of type I collagen and inflammatory cytokines by peripheral blood fibrocytes. J Immunol 1998, 160:419–425. 738 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes Kuwana M, Okazaki Y, Kodama H, et al.: Human circulating CD14+ monocytes as a source of progenitors that exhibit mesenchymal cell differentiation. J Leukoc Biol 2003, 74:833–845. This paper describes MOMPS as having features of several different mesenchymal cell types. 28 • 29 Hashimoto N, Jin H, Liu T, et al.: Bone marrow-derived progenitor cells in pulmonary fibrosis. J Clin Invest 2004, 113:243–252. •• This is an interesting paper that uses GFP labeled bone marrow chimera mice to induce pulmonary fibrosis secondary to bleomycin inhalation. They found that the bone marrow contributes fibroblasts to the fibrotic process in lungs of the bleomycin-treated mice. 30 Pilling D, Buckley CD, Salmon M, et al.: Inhibition of fibrocyte differentiation by serum amyloid P1. J Immunol 2003, 171:5537–5546. •• Excellent demonstration that serum amyloid protein inhibits fibrocyte development and that SSc sera have low levels of serum amyloid protein. These low levels are postulated to possibly predispose SSc monocytes to transdifferentiate into fibrocytes. Recent advances in fibroblast signaling and biology in scleroderma Jaspreet Pannu and Maria Trojanowska Purpose of review Systemic sclerosis is a complex disease manifesting itself by fibrosis of skin and other internal organs. Fibroblasts isolated from scleroderma lesions and cultured in vitro are characterized by increased synthesis of collagen and other extracellular matrix proteins, consistent with the disease phenotype. Cultured systemic sclerosis fibroblasts therefore serve as a principal experimental model for studying the molecular and cellular mechanisms involved in collagen overproduction in this disease. This review will discuss recent findings related to intracellular signal transduction pathways implicated in deregulated extracellular matrix deposition by systemic sclerosis fibroblasts. Recent findings Recent findings suggest that constitutively elevated synthesis of extracellular matrix by cultured systemic sclerosis fibroblasts is, at least in part, due to the aberrant activation of the autocrine transforming growth factor- signaling. Enhanced constitutive transforming growth factor- signaling may result from the elevated levels of transforming growth factor- receptor type I and/or inappropriate activation of Smad3. These alterations of the transforming growth factor- signaling in systemic sclerosis fibroblasts may facilitate increased collagen production in vivo even under conditions of low ligand availability. However, there exist many inconsistencies among published reports regarding the detailed mechanisms of this pathway in systemic sclerosis fibroblasts, and additional studies in this area are needed. Other signaling molecules implicated in fibrotic phenotype include several members of the protein kinase C family, mammalian target of rapamycin, mitogen-activated protein kinase, necdin, reactive oxygen species, and sphingolipids. These signaling pathways may work in conjunction with transforming growth factor- signaling to regulate the behavior of systemic sclerosis fibroblasts. Summary Alterations in multiple signaling pathways contribute to elevated extracellular matrix synthesis by systemic sclerosis fibroblasts. Improved understanding of the key signaling molecules may provide a novel avenue for therapeutic interventions. Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA Correspondence to Maria Trojanowska, Medical University of South Carolina, Division of Rheumatology and Immunology, 96 Jonathan Lucas Street, Suite 912, Charleston, SC 29425, USA Tel: 843 792 7921; fax: 843 792 7121; e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:739–745 Abbreviations ECM MAPK mTOR PKC SSc TGF extracellular matrix mitogen-activated protein kinase mammalian target of rapamycin protein kinase C systemic sclerosis, scleroderma transforming growth factor © 2004 Lippincott Williams & Wilkins 1040–8711 Introduction Fibroblasts cultured from the skin or lungs of patients with systemic sclerosis (scleroderma, SSc) show elevated collagen synthesis and several other phenotypic differences when compared with healthy skin fibroblasts [1•, 2]. Earlier findings indicated that the collagen type I gene is upregulated at the transcriptional level in SSc fibroblasts [3,4]. These findings have been facilitated by the cloning of the regulatory proximal regions of the collagen type I genes [5]. These fruitful studies resulted in characterization of the transcription factors altered in SSc fibroblasts and have been summarized in several recent reviews [6–8]. In comparison, the signal transduction pathways contributing to SSc in general, and collagen synthesis in particular, until recently, have been largely unexplored. This review will discuss our current knowledge of the signaling molecules involved in the regulation of cell behavior relevant to fibrosis with the emphasis on those pathways that are deregulated in SSc fibroblasts (Table 1). As outlined below, significant progress has been made in characterizing SSc-specific alterations of the transforming growth factor (TGF)- signaling cascade. The TGF- signaling pathway is discussed in detail elsewhere, and readers are referred to recent reviews on this topic [9,10,11•]. Transforming growth factor- signaling Keywords scleroderma, systemic sclerosis, fibroblasts, signaling pathways Curr Opin Rheumatol 16:739–745. © 2004 Lippincott Williams & Wilkins. The TGF- is the most potent fibrogenic cytokine and is considered to play a principal role in various fibrotic diseases, including SSc [9]. Accumulated evidence suggests that defected TGF- signaling contributes to the phenotypic alterations of SSc fibroblasts in culture. This 739 740 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes area of investigation, however, will require further clarification because different laboratories have reported inconsistent findings. The differences may reflect heterogeneity of the disease, ethnic background, or stage of the disease. Elevated levels of transforming growth factor- receptors contribute to phenotypic changes in systemic sclerosis fibroblasts: the hypothesis Cultured SSc fibroblasts differ from healthy skin fibroblasts with regard to extracellular matrix (ECM) production and other cellular responses related to fibrosis [12]. Furthermore, many of the characteristics of SSc fibroblasts can be induced in healthy skin fibroblasts by TGF- treatment [13]. It was hypothesized that altered phenotype of SSc fibroblast in culture is dependent on autocrine TGF- signaling. Because TGF- production has been shown to be similar in SSc and healthy skin fibroblasts, whereas TGF- receptor levels were shown to be elevated in SSc fibroblasts, it was suggested that the elevated levels of TGF- receptors is the primary defect leading to enhanced autocrine TGF- signaling [14,15]. Although this hypothesis has been supported by subsequent studies, the finding of the elevated TGF- receptor levels has not been universally reproduced. Work by Ihn et al. [16], Kubo et al. [17], and Yamane et al. [18] consistently demonstrated elevated levels of TGF- receptor-I and -II in Japanese patients. However, in a recent study by Pannu et al. [19••], only TGF- receptor-I protein levels were shown to be consistently elevated in SSc fibroblasts, whereas TGF- receptor-II levels were only occasionally increased. In a majority of SSc fibroblasts analyzed in the latter study, TGF- receptor-II protein levels were modestly decreased. Interestingly, decreased TGF- receptor-II levels were observed in fibroblasts obtained from Caucasian patients, whereas in African-American patients, TGF- receptorII levels were either elevated or not changed (Pannu and Trojanowska, unpublished observations). This intriguing preliminary observation warrants further investigation. A study by Dong et al. [20] found no differences in TGF- receptor levels. A marked heterogeneity with regard to TGF- receptor-I expression in vivo was observed in a study by Pannu et al. [19••], which may also explain some of the discrepancies among different reports. The evidence for autocrine transforming growth factor- signaling in systemic sclerosis fibroblasts Ihn et al. have shown that blockade of endogenous TGF- signaling via neutralizing antibodies or antisense TGF- oligonucleotides prevented upregulated collagen synthesis by SSc fibroblasts [16]. Blockade of TGF- signaling also reduced the expression of Smad7, another TGF- inducible gene, upregulated in SSc fibroblasts [21••]. Surprisingly, upregulation of collagen synthesis by a subset of SSc fibroblasts was resistant to a blockade of TGF- signaling by a different approach, ie, the overexpression of the kinase-deficient TGF- receptor-II [19••]. Pannu et al. [19••] have shown that SSc fibroblasts with the highest levels of TGF- receptor-I were the least responsive, suggesting that upregulation of collagen synthesis by SSc fibroblasts may primarily depend on the signaling downstream from the TGF- receptor-I. In support of this concept, it was shown that forced expression of TGF- receptor-I (but not receptor-II) in healthy fibroblasts in a range corresponding to the elevated levels of TGF- receptor-I found in SSc fibroblasts results in elevated collagen synthesis. Of note, studies from other experimental models showed that decreased signaling from TGF- receptor-II correlated with the induction of epithelial mesenchymal transition and preservation of ECM induction with the selective loss of growth inhibitory responses [22,23]. Furthermore, in transgenic mice with fibroblast-specific expression of a kinase-deficient TGF- receptor-II receptor dermal and pulmonary fibrosis developed, suggesting that perturbations of specific aspects of the TGF- signaling pathway in fibroblasts may induce fibrosis in vivo [24]. Transforming growth factor- receptor turnover is defective in systemic sclerosis fibroblasts What is the mechanism of upregulation of TGF- receptors in SSc fibroblasts? According to the current theory of TGF- receptor trafficking, receptor internalization via the caveolae pathway leads to receptor degradation, whereas the clathrin-dependent internalization into SARA-containing early endosomes promotes postreceptor signaling [25••]. Interestingly, the half-life of TGF- receptor-I protein is significantly longer than the half-life of TGF- receptor-II protein, suggesting distinct degradation pathways for these two receptors [26]. Furthermore, -arrestin 2 has been reported to be involved in the co-internalization of TGF- receptor-II with TGF- receptor-III but not TGF- receptor-I, thus strengthening the viewpoint of different internalization/degradation pathways for these receptors [27]. Significantly, a recent study by Asano et al. [21••] revealed that TGF- receptor-I protein is stabilized in SSc fibroblasts. These authors also showed that in SSc fibroblasts, TGF- receptors are localized to presently uncharacterized subcellular structures with a punctate appearance. Because TGF- signaling appears to be constitutively activated in SSc cells, presumably these receptor-containing structures represent early endosomes in which signaling occurs. Paradoxically, however, Smad7 was colocalized with TGF- receptors to these subcellular structures. In addition, it was shown that in SSc fibroblasts, TGF- receptor-I turnover was insensitive to the ectopically expressed Smurf1/2, whereas such treatment promoted receptor degradation in control fibroblasts [21••]. Because the association of TGF- receptors with Smad7- Fibroblast signaling and biology in scleroderma Pannu and Trojanowska 741 Table 1. Signaling molecules implicated in scleroderma Molecule Functional relevance Status in SSc fibroblasts Study TGF- pathway: receptor type I Main signaling receptor in TGF pathway Elevated levels in vitro and in vivo Receptor type II Binds ligand and activates TGFRI Nonsignaling high-affinity TGF receptor generally associated with endothelial cells Downstream effectors of TGF signaling pathway Elevated or decreased levels in vitro; elevated in vivo Elevated levels in vitro Kawakami et al. [15], Kubo et al. [17], Yamane et al. [18], Pannu et al. [19••] Kawakami et al. [15], Yamane et al. [18], Pannu et al. [19••] Leask et al. [61] Receptor type III (endoglin/CD1 05) Smad2/3 Smad7 Inhibits TGF signaling ␣11 and ␣21 integrins ␣v5 integrin Cell surface receptors for collagen Receptor for vitronectin— stimulates human alpha2(I) collagen promoter activity through Sp-1 and Smad3 Receptors for endothelin-1; promote myofibroblast phenotype in lung fibroblasts Endothelin receptor A and B Angiotensin II receptor type I (AT1) PDGF receptor ␣ PDGF receptor  Necdin PKC␦ PKC p38 MAPK JNK/SAPK ERK MAPK Cell surface receptor for angiotensin II (Ang II). Signaling receptor for PDGF A, B, and C; implicated in lung and cardiac fibrosis Signaling receptor for PDGF B and D Nuclear protein; associates with pre-IL-1␣ and stimulates collagen production Member of the novel subfamily of PKCs implicated in collagen and fibronectin upregulation Member of the conventional subfamily of PKCs implicated TN-C upregulation and promotion of myofibroblast phenotype Mediator of TGF- induced collagen synthesis Negative regulator of collagen transcription Implicated in CTGF and collagen upregulation Elevated expression and phosphorylation levels; constitutive nuclear localization Conflicting data: elevated and decreased levels Unchanged Asano et al. [63••} Elevated total ET-1 receptor; decreased ETA and increased ETB levels in SSc lung fibroblasts Elevated levels in vitro and in vivo Elevated levels in vitro Abraham et al. [64], Shi-Wen et al. [65] Elevated in vivo Klareskog et al. [70] Both, necdin and pre-IL-1␣ have elevated expression Kawaguchi et al. [71], Hu et al. [72••] Elevated levels in vitro; interacts with TGF pathway in pulmonary fibrosis Decreased levels in SSC lung fibroblasts; constitutively associated with ␣-SMA Jimenez et al. [49], Gore-Hyer et al. [50], Zhang et al. [52], Mimura et al. [73] Tourkina et al. [54], Tourkina et al. [55] No change Sato et al. [74] Unknown Fisher et al. [75] Constitutively activated in SSc lung and dermal fibroblasts Tourkina, unpublished observations; Pannu and Trojanowska, unpublished observations Shegogue et al. [59••]; Pannu and Trojanowska, unpublished observations Yamanaka and Trojanowska, unpublished observations Sato et al. [76] Positive regulator of collagen synthesis Elevated levels in vitro Sphingosine kinase Upregulated by TGF; mediates stimulation of TIMP-1 Induced by TGF-; cooperates with TGF- in collagen upregulation Implicated in stimulation of collagen synthesis Unknown Reactive oxygen species Dong et al. [20], Asano et al. [21••], Mori et al. [30] Herzhoff et al. [62] Elevated levels in vitro and in vivo mTOR Ceramide Mori et al. [30••], Asano et al. [31] Unknown Elevated levels in SSc fibroblasts Kawaguchi et al. [66•] Yamakage et al. [67], Zhuo et al. [68], Ponten et al. [69] Sambo et al. [77] IL, interleukin; TGF, transforming growth factor; MAPK, mitogen-activated protein kinase; mTOR, mammalian target of rapamycin. Smurf2 is linked to the caveolae-dependent degradation pathway, it appears that in SSc fibroblasts a receptor degradation defect occurs downstream from Smad7/Smurf2/TGF- receptor complex formation. At present, it is difficult to reconcile the existing observations from SSc fibroblasts with the current theories re- garding TGF- receptor internalization and turnover. Clearly, additional studies are needed to characterize the vesicles involved in TGF- signaling in SSc cells. It may be relevant that fibroblasts from SSc lung tissues have lower levels of caveolin-1 (Tourkina, unpublished data). If this observation extends to skin fibroblasts, 742 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes it may suggest that the balance between the caveolaedependent degradation pathway and clathrin-dependent signaling pathway is shifted toward the clathrin-dependent pathway in SSc fibroblasts (Fig. 1). Smad2/3 pathway is constitutively activated in systemic sclerosis fibroblasts R-Smads are the primary transducers of TGF- signaling. Their role in fibrosis has also been confirmed in various animal models [28•,29]. Thus, it is reasonable to expect that constitutive activation of TGF- signaling in SSc fibroblasts will also involve R-Smad activation. Recent studies by Mori et al. [30••] support this concept. Their study showed elevated phosphorylation levels of Smad2 and increased nuclear localization of Smad2/3 in SSc fibroblasts. Consistent with the study by Pannu et al. [19••], nuclear localization of Smads in SSc fibroblasts was insensitive to the blockade of TGF- signaling via neutralizing TGF- antibody or overexpression of kinase-deficient TGF- receptor-II [30••]. Other investigators reported moderately increased levels of phosphorylated Smad2 and Smad3 in SSc fibroblasts [21••,31]. A recent study by Asano et al. [31] showed that a treatment with the pharmacologic inhibitor of PI-3 kinase abrogated constitutive Smad3 phosphorylation in SSc fibroblasts. This finding is consistent with the previous study, which showed that inhibitors of the PI-3 kinase pathway markedly reduced the expression of TGF- receptor-II Figure 1. Hypothetical model of alterations in transforming growth factor (TGF)- signaling pathway occurring in systemic sclerosis (SSc) fibroblasts in SSc fibroblasts [32]. Therefore, although it is formally possible that Smad2/3 phosphorylation in SSc fibroblasts is independent of TGF- signaling, existing evidence places the activation of Smad2/3 downstream from the TGF- receptors. Clearly, more studies are needed to explain the mechanism involved in constitutive phosphorylation and nuclear localization of Smads in SSc fibroblasts, especially in view of recent findings on Smad regulation. It is now evident that Smads actively shuttle between the nucleus and the cytoplasm and that the subcellular localization of Smads is subjected to a complex regulatory mechanism that is not yet fully elucidated [33]. In contrast to consistent findings of elevated phosphorylation status of Smad3 in SSc fibroblasts, reports regarding the expression levels of Smad7 are highly inconsistent. Asano et al. [21••] reported elevated levels of Smad7 in SSc fibroblasts, whereas no consistent changes in expression levels of Smad7 were reported by other studies [30••] . Finally, reduced Smad7 expression in SSc fibroblasts was reported by Dong et al. [20]. Activation of the Smad pathway cannot, however, explain some of the principal characteristics of SSc fibroblasts. For example, elevated expression of CTGF in SSc fibroblasts was shown to be Smad-independent [34]. A possible explanation may be provided by our recent unpublished studies. Dermal fibroblasts with ectopically increased levels of TGF- receptor type I, as described by Pannu et al. [19••], showed elevated mRNA expression of COL1A1, COL1A2, and CTGF. Furthermore, elevated TGF- receptor-I levels resulted in activation of ERK mitogen-activated protein kinase (MAPK) pathway, without activation of the Smad2/3 pathway (Pannu and Trojanowska, unpublished observations). These preliminary observations suggest a TGF-–dependent, Smad-independent upregulation of CTGF in SSc fibroblasts, which involves activation of ERKs. Regulation of CTGF expression by an ERK-dependent pathway has previously been reported [35,36••]. Does transforming growth factor- play a role in maintenance of fibrosis? In SSc fibroblasts increased receptor levels (either R1 or both) lead to constitutive activation of Smad2/3 pathway and upregulation of matrix genes. Other factors, presently uncharacterized, may also contribute to Smad2/3 activation. The downregulation of the TGF- signaling pathway is impaired in SSc fibroblasts, possibly owing to the defects in the components of caveolae, including caveolin and/or sphingolipids. Data from Yamane et al. [18], Pannu et al. [19••], Varga [9], and Asano et al. [21••]. Early SSc lesions are characterized by the influx of TGF–positive cells. This elevated expression of TGF- is limited to the inflammatory leading edge, and TGF- is no longer detectable during subsequent stages of the disease, which is characterized by elevated ECM deposition [37]. On the basis of these observations, it has been suggested that TGF- signaling is involved in the early stages, whereas other signaling pathways (eg, CTGF) are responsible for ECM deposition during chronic stages of the disease [38]. The question, then, is whether TGF- signaling is still active in fibroblasts in vivo in these later stages and whether it contributes to the progression of the disease. Although involvement of CTGF is critical Fibroblast signaling and biology in scleroderma Pannu and Trojanowska 743 for the fibrogenic effects of TGF-, we propose that TGF- is the major player also in chronic stages of SSc [39]. First, it cannot be excluded that the low levels of TGF- are synthesized by fibroblasts in the skin in vivo. Furthermore, latent TGF- sequestered by ECM may serve as an additional source of the ligand. Consistent with this possibility, it has been observed that fibrillincontaining fibrils are unstable in SSc skin in vivo, which may lead to the release of the matrix-bound TGF- [40]. Evidence from cultured SSc fibroblasts, as well as the demonstration of elevated TGF- receptor levels in vivo, strongly suggests that specific alterations of the TGF- signaling may facilitate robust ECM synthesis in vivo even when the ligand availability is low [17,19••]. Although additional studies are needed to support this notion, it is noteworthy that those alterations were also observed in collagen-producing cells isolated from other fibrotic tissues [41–44]. The question remains how SSc fibroblasts acquire these changes. Recent studies of animal models of fibrosis strongly suggest that the collagenproducing fibroblasts seen in the fibrotic lesions either originate in the circulation or are derived from epithelial cells via epithelial-mesenchymal transformation (EMT) [45,46]. This possibility remains to be tested for SSc skin and lung fibroblasts. Other possible mechanisms to explain the phenotypic differences of SSc fibroblasts include mutational changes and gene polymorphisms [47]. Protein kinase C pathway The protein kinase C (PKC) isoforms constitute a family of serine-threonine kinases that form three subfamilies based on structural homology and sensitivity to activators and have been extensively reviewed [48]. Elevated levels of PKC␦ were found in SSc skin fibroblasts [49]. Furthermore, blockade of the PKC␦ activity by a specific inhibitor, rottlerin, significantly reduced collagen synthesis in SSc and healthy fibroblasts [49]. PKC␦ activity was also necessary to mediate the stimulatory effect of CTGF in cooperation with insulin/IGF-I signaling on collagen synthesis by SSc fibroblasts [50]. Importantly, PKC␦ has been shown to interact with the TGF- signaling pathway (Fig. 2). In mesangial cells, TGF- activated PKC␦ and, in turn, PKC␦, positively regulated Smad3 transcription activity and COL1A2 transcription [51]. Likewise, TGF- activated PKC␦ in lung fibroblasts, whereas interleukin-7, an antagonist of TGF- function, inhibited PKC␦ activity [52]. Another PKC isoform, PKC, has been reported to function aberrantly in SSc lung fibroblasts. PKC activity has been linked to myofibroblast differentiation, upregulation of Tenascin C expression, and increased sensitization of SSc lung fibroblasts to proapoptotic agents [53–55]. Figure 2. Crosstalk between transforming growth factor (TGF)- and other signaling pathways in fibrosis TGF- activates PKC␦, which contributes to Smad-mediated upregulation of collagen gene. TGF-–dependent interaction between ␣v3 integrin (Tenascin-C or Vitronectin receptor) and TRII enhances the proliferative effects of TGF-. ␣v3 also activates the mTOR pathway, which positively regulates collagen transcription and mRNA stability. mTOR is an effector of the Akt pathway, which was shown to be constitutively activated in SSc fibroblasts. There is evidence that mTOR phosphorylates PKC␦. PKC␦, vitronectin receptor (␣v5), and mTOR levels are upregulated in SSc fibroblasts. This crosstalk may contribute to enhanced collagen production by SSc fibroblasts. Data from Runyan et al. [51], Shegogue et al. [59••], Maeshima et al. [79], Jun et al. [80], Parekh et al. [81], Jimenez et al. [49], and Asano et al. [63••]. tion [56]. Because mTOR is a key nutrient sensor as well as a mediator of mitogen and hormone signaling through the PI3 kinase/Akt pathway, it is postulated that it functions as an integrator of pathways regulating cell size and cell division [57,58]. mTOR has been shown to positively regulate collagen production in dermal fibroblasts via a P1-3-kinase–independent pathway [59••]. Interestingly, COL1A1 and COL1A2 are differentially regulated by mTOR. COL1A1 is regulated at transcriptional and posttranscriptional (mRNA stability) levels, whereas COL1A2 is regulated via mRNA stability only. The relevance of this differential regulation is currently unknown. Our preliminary studies indicate that mTOR protein levels are elevated in SSc fibroblasts (Pannu and Trojanowska, unpublished observations). How the mTOR pathway is integrated with other signaling pathways involved in collagen synthesis is not known; however, functional interactions between mTOR and PKC␦ have been reported (Fig. 2) [60]. Mammalian target of rapamycin pathway Conclusion The mammalian target of rapamycin (mTOR), a member of the PI3 kinase superfamily, plays a key role in the regulation of cell growth, proliferation, and differentia- The regulation of ECM synthesis occurs at many levels and involves integration of the signals from the cytokines, various matrix molecules, and other cells. Delin- 744 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes eation of the signaling molecules involved in this process and especially the key molecules that are deregulated in SSc fibroblasts is an area of intense research. Many parallels between SSc fibroblasts and collagen-producing cells from other fibrotic diseases have been found. The signaling molecules, including components of the TGF- signaling cascade, PKC␦, or mTOR, may serve as universal targets for designing therapies to benefit patients with SSc and other fibrotic diseases. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest Jimenez SA, Derk CT: Following the molecular pathways toward an understanding of the pathogenesis of systemic sclerosis. Ann Intern Med 2004, 140:37–50. A comprehensive review of the pathogenesis of SSc, encompassing current knowledge of causative agents and physiologic alterations that culminate in fibrosis. Pannu J, Gore-Hyer E, Yamanaka M, et al.: An increased transforming growth factor beta receptor type I:type II ratio contributes to elevated collagen protein synthesis that is resistant to inhibition via a kinase-deficient transforming growth factor beta receptor type II in scleroderma. Arthritis Rheum 2004, 50:1566–1577. This study demonstrates the functional significance of the elevated TGF- receptor-I in SSc fibroblasts. 19 •• 20 Asano Y, Ihn H, Yamane K, et al.: Impaired Smad7-Smurf-mediated negative regulation of TGF-beta signaling in scleroderma fibroblasts. J Clin Invest 2004, 113:253–264. This important study shows that TRI protein is stabilized in SSc fibroblasts. It also demonstrates that Smad7-Smurf2-mediated degradation of TGF- receptors is impaired in SSc fibroblasts. 21 •• 22 Oft M, Heider KH, Beug H: TGFbeta signaling is necessary for carcinoma cell invasiveness and metastasis. Curr Biol 1998, 8:1243–1252. 23 Chen RH, Ebner R, Derynck R: Inactivation of the type II receptor reveals two receptor pathways for the diverse TGF-beta activities. Science 1993, 260: 1335–1338. 24 Denton CP, Zheng B, Evans LA, et al.: Fibroblast-specific expression of a kinase-deficient type II transforming growth factor beta (TGFbeta) receptor leads to paradoxical activation of TGFbeta signaling pathways with fibrosis in transgenic mice. J Biol Chem 2003, 278:25109–25119. 1 • 2 LeRoy EC: A brief overview of the pathogenesis of scleroderma (systemic sclerosis). Ann Rheum Dis 1992, 51:286–288. 3 Kikuchi K, Hartl CW, Smith EA, et al.: Direct demonstration of transcriptional activation of collagen gene expression in systemic sclerosis fibroblasts: insensitivity to TGF beta 1 stimulation. Biochem Biophys Res Commun 1992, 187:45–50. 4 Hitraya EG, Jimenez SA: Transcriptional activation of the alpha 1(I) procollagen gene in systemic sclerosis dermal fibroblasts: role of intronic sequences. Arthritis Rheum 1996, 39:1347–1354. 5 Boast S, Su MW, Ramirez F, et al.: Functional analysis of cis-acting DNA sequences controlling transcription of the human type I collagen genes. J Biol Chem 1990, 265:13351–13356. 6 Trojanowska M: Molecular aspects of scleroderma. Front Biosci 2002, 7:d608–d618. 7 Ghosh AK: Factors involved in the regulation of type I collagen gene expression: implication in fibrosis. Exp Biol Med (Maywood) 2002, 227:301–314. 8 Jimenez SA, Saitta B: Alterations in the regulation of expression of the alpha 1(I) collagen gene (COL1A1) in systemic sclerosis (scleroderma). Springer Semin Immunopathol 1999, 21:397–414. 9 Varga J: Scleroderma and Smads: dysfunctional Smad family dynamics culminating in fibrosis. Arthritis Rheum 2002, 46:1703–1713. 10 Shi Y, Massague J: Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell 2003, 113:685–700. 11 Dijke Pt P, Hill CS: New insights into TGF-beta-Smad signalling. Trends Biochem Sci 2004, 29:265–273. • A review of newly identified regulatory mechanisms that determine the signal specificity, duration, and intensity of the various events in TGF- signaling. 12 Strehlow D, Korn JH: Biology of the scleroderma fibroblast. Curr Opin Rheumatol 1998, 10:572–578. 13 Massague J: TGF-beta signal transduction. Annu Rev Biochem 1998, 67:753–791. 14 Needleman BW, Choi J, Burrows-Mezu A, Fontana JA: Secretion and binding of transforming growth factor beta by scleroderma and normal dermal fibroblasts. Arthritis Rheum 1990, 33:650–656. 15 Kawakami T, Ihn H, Xu W, et al.: Increased expression of TGF-beta receptors by scleroderma fibroblasts: evidence for contribution of autocrine TGF-beta signaling to scleroderma phenotype. J Invest Dermatol 1998, 110:47–51. 16 Ihn H, Yamane K, Kubo M, Tamaki K: Blockade of endogenous transforming growth factor beta signaling prevents up-regulated collagen synthesis in scleroderma fibroblasts: association with increased expression of transforming growth factor beta receptors. Arthritis Rheum 2001, 44:474–480. 17 Kubo M, Ihn H, Yamane K, Tamaki K: Upregulated expression of transforming growth factor-beta receptors in dermal fibroblasts of skin sections from patients with systemic sclerosis. J Rheumatol 2002, 29:2558–2564. 18 Yamane K, Ihn H, Kubo M, Tamaki K: Increased transcriptional activities of transforming growth factor beta receptors in scleroderma fibroblasts. Arthritis Rheum 2002, 46:2421–2428. Dong C, Zhu S, Wang T, et al.: Deficient Smad7 expression: a putative molecular defect in scleroderma. Proc Natl Acad Sci USA 2002, 99:3908– 3913. Di Guglielmo GM, Le Roy C, Goodfellow AF, et al.: Distinct endocytic pathways regulate TGF-beta receptor signalling and turnover. Nat Cell Biol 2003, 5:410–421. This pivotal study defines the two main pathways that regulate endocytic trafficking of TGF- receptors. The authors show that clathrin-dependent internalization triggers signaling response, whereas lipid raft-caveolar internalization mediates ubiquitin-dependent degradation. 25 •• 26 Koli KM, Arteaga CL: Processing of the transforming growth factor beta type I and II receptors: biosynthesis and ligand-induced regulation. J Biol Chem 1997, 272:6423–6427. 27 Chen W, Kirkbride KC, How T, et al.: Beta-arrestin 2 mediates endocytosis of type III TGF-beta receptor and down-regulation of its signaling. Science 2003, 301:1394–1397. 28 Flanders KC: Smad3 as a mediator of the fibrotic response. Int J Exp Pathol 2004, 85:47–64. • A very informative review of the role of Smad3 in fibrosis of various organs. 29 Lakos G, Chen SJ, Ferreira AM, et al.: Targeted disruption of TGF-/Smad3 signaling modulates skin fibrosis in a mouse model of scleroderma. Am J Physiol 2004, in press. Mori Y, Chen SJ, Varga J: Expression and regulation of intracellular SMAD signaling in scleroderma skin fibroblasts. Arthritis Rheum 2003, 48:1964– 1978. This study shows for the first time that Smad2/3 are constitutively activated in SSc fibroblasts. 30 •• 31 Asano Y, Ihn H, Yamane K, et al.: Phosphatidylinositol 3-kinase is involved in alpha2(I) collagen gene expression in normal and scleroderma fibroblasts. J Immunol 2004, 172:7123–7135. 32 Yamane K, Ihn H, Tamaki K: Epidermal growth factor up-regulates expression of transforming growth factor beta receptor type II in human dermal fibroblasts by phosphoinositide 3-kinase/Akt signaling pathway: resistance to epidermal growth factor stimulation in scleroderma fibroblasts. Arthritis Rheum 2003, 48:1652–1666. 33 Reguly T, Wrana JL: In or out? The dynamics of Smad nucleocytoplasmic shuttling. Trends Cell Biol 2003, 13:216–220. 34 Holmes A, Abraham DJ, Sa S, et al.: CTGF and SMADs: maintenance of scleroderma phenotype is independent of SMAD signaling. J Biol Chem 2001, 276:10594–10601. 35 Chen Y, Blom IE, Sa S, et al.: CTGF expression in mesangial cells: involvement of SMADs, MAP kinase, and PKC. Kidney Int 2002, 62:1149–1159. 36 Leask A, Abraham DJ: TGF-beta signaling and the fibrotic response. FASEB J 2004, 18:816–827. •• This comprehensive review discusses the current state of knowledge on profibrotic (TGF-, CTGF, ED-A FN) and antifibrotic (TNF␣ and INF␥) pathways. 37 Querfeld C, Eckes B, Huerkamp C, et al.: Expression of TGF-beta 1, -beta 2 and -beta 3 in localized and systemic scleroderma. J Dermatol Sci 1999, 21: 13–22. Fibroblast signaling and biology in scleroderma Pannu and Trojanowska 745 38 Leask A, Holmes A, Abraham DJ: Connective tissue growth factor: a new and important player in the pathogenesis of fibrosis. Curr Rheumatol Rep 2002, 4:136–142. 62 39 Grotendorst GR: Connective tissue growth factor: a mediator of TGF-beta action on fibroblasts. Cytokine Growth Factor Rev 1997, 8:171–179. 40 Wallis DD, Tan FK, Kielty CM, et al.: Abnormalities in fibrillin 1-containing microfibrils in dermal fibroblast cultures from patients with systemic sclerosis (scleroderma). Arthritis Rheum 2001, 44:1855–1864. 63 Asano Y, Ihn H, Yamane K, et al.: Increased expression levels of integrin alphavbeta5 on scleroderma fibroblasts. Am J Pathol 2004, 164:1275–1292. •• This study demonstrates elevated expression levels of vitronectin receptor, ␣v5, in SSc fibroblasts in vitro and in vivo. This study also shows that ectopic expression of 5 in dermal fibroblasts leads to elevated collagen promoter activity and increased phosphorylation of Smad3. 41 Inagaki Y, Mamura M, Kanamaru Y, et al.: Constitutive phosphorylation and nuclear localization of Smad3 are correlated with increased collagen gene transcription in activated hepatic stellate cells. J Cell Physiol 2001, 187:117– 123. 42 Tahashi Y, Matsuzaki K, Date M, et al.: Differential regulation of TGF-beta signal in hepatic stellate cells between acute and chronic rat liver injury. Hepatology 2002, 35:49–61. 43 Baer HU, Friess H, Abou-Shady M, et al.: Transforming growth factor betas and their receptors in human liver cirrhosis. Eur J Gastroenterol Hepatol 1998, 10:1031–1039. Herzhoff K, Sollberg S, Huerkamp C, et al.: Fibroblast expression of collagen integrin receptors alpha1beta1 and alpha2beta1 is not changed in systemic scleroderma. Br J Dermatol 1999, 141:218–223. 64 Abraham DJ, Vancheeswaran R, Dashwood MR, et al.: Increased levels of endothelin-1 and differential endothelin type A and B receptor expression in scleroderma-associated fibrotic lung disease. Am J Pathol 1997, 151:831– 841. 65 Shi-Wen X, Chen Y, Denton CP, et al.: Endothelin-1 promotes myofibroblast induction through the ETA receptor via a rac/phosphoinositide 3-kinase/Aktdependent pathway and is essential for the enhanced contractile phenotype of fibrotic fibroblasts. Mol Biol Cell 2004, 15:2707–2719. Kawaguchi Y, Takagi K, Hara M, et al.: Angiotensin II in the lesional skin of systemic sclerosis patients contributes to tissue fibrosis via angiotensin II type 1 receptors. Arthritis Rheum 2004, 50:216–226. First report demonstrating elevated angiotensin II and angiotensin receptors in fibroblasts and skin sections from diffuse cutaneous SSc skin. 66 • 44 McCaffrey TA: TGF-betas and TGF-beta receptors in atherosclerosis. Cytokine Growth Factor Rev 2000, 11:103–114. 45 Hashimoto N, Jin H, Liu T, et al.: Bone marrow-derived progenitor cells in pulmonary fibrosis. J Clin Invest 2004, 113:243–252. 46 Kalluri R, Neilson EG: Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 2003, 112:1776–1784. 47 Ahmed SS, Tan FK: Identification of novel targets in scleroderma: update on population studies, cDNA arrays, SNP analysis, and mutations. Curr Opin Rheumatol 2003, 15:766–771. 48 Newton AC: The ins and outs of protein kinase C. Methods Mol Biol 2003, 233:3–7. 49 Jimenez SA, Gaidarova S, Saitta B, et al.: Role of protein kinase C-delta in the regulation of collagen gene expression in scleroderma fibroblasts. J Clin Invest 2001, 108:1395–1403. 50 Gore-Hyer E, Pannu J, Smith EA, et al.: Selective stimulation of collagen synthesis in the presence of costimulatory insulin signaling by connective tissue growth factor in scleroderma fibroblasts. Arthritis Rheum 2003, 48:798– 806. 51 Runyan CE, Schnaper HW, Poncelet AC: Smad3 and PKCdelta mediate TGF-beta1-induced collagen I expression in human mesangial cells. Am J Physiol Renal Physiol 2003, 285:F413–F422. 52 Zhang L, Keane MP, Zhu LX, et al.: IL-7 and TGF-beta play counter-regulatory roles in PKC-delta-dependent control of fibroblast collagen synthesis in pulmonary fibrosis. J Biol Chem 2004, 279:28315–28319. 73 53 Bogatkevich GS, Tourkina E, Abrams CS, et al.: Contractile activity and smooth muscle alpha-actin organization in thrombin-induced human lung myofibroblasts. Am J Physiol Lung Cell Mol Physiol 2003, 285:L334–L343. Mimura Y, Ihn H, Jinnin M, et al.: Epidermal growth factor induces fibronectin expression in human dermal fibroblasts via protein kinase C delta signaling pathway. J Invest Dermatol 2004, 122:1390–1398. 74 54 Tourkina E, Hoffman S, Fenton JW 2nd, et al.: Depletion of protein kinase C epsilon in normal and scleroderma lung fibroblasts has opposite effects on tenascin expression. Arthritis Rheum 2001, 44:1370–1381. Sato M, Shegogue D, Gore EA, et al.: Role of p38 MAPK in transforming growth factor beta stimulation of collagen production by scleroderma and healthy dermal fibroblasts. J Invest Dermatol 2002, 118:704–711. 75 55 Tourkina E, Gooz P, Oates JC, et al.: Curcumin-induced apoptosis in scleroderma lung fibroblasts: role of protein kinase C {epsilon}. Am J Respir Cell Mol Biol 2004, 31:28–35. Fisher GJ, Datta S, Wang Z, et al.: c-Jun-dependent inhibition of cutaneous procollagen transcription following ultraviolet irradiation is reversed by alltrans retinoic acid. J Clin Invest 2000, 106:663–670. 76 56 Harris TE, Lawrence JC, Jr. TOR signaling. Sci STKE 2003;2003(212):re15. Sato M, Markiewicz M, Yamanaka M, et al.: Modulation of transforming growth factor-beta (TGF-beta) signaling by endogenous sphingolipid mediators. J Biol Chem 2003, 278:9276–9282. 57 Kuruvilla FG, Schreiber SL: The PIK-related kinases intercept conventional signaling pathways. Chem Biol 1999, 6:R129–R136. 77 58 Abraham RT: mTOR as a positive regulator of tumor cell responses to hypoxia. Curr Top Microbiol Immunol 2004, 279:299–319. Sambo P, Baroni SS, Luchetti M, et al.: Oxidative stress in scleroderma: maintenance of scleroderma fibroblast phenotype by the constitutive upregulation of reactive oxygen species generation through the NADPH oxidase complex pathway. Arthritis Rheum 2001, 44:2653–2664. 78 Scaffidi AK, Petrovic N, Moodley YP, et al.: Alpha vbeta 3 integrin interacts with the TGFbeta type II receptor to potentiate the proliferative effects of TGFbeta 1 in living human lung fibroblasts. J Biol Chem 2004, Jun 8 (e-pub ahead of print). 79 Maeshima Y, Sudhakar A, Lively JC, et al.: Tumstatin, an endothelial cellspecific inhibitor of protein synthesis. Science 2002, 295:140–143. 80 Jun JB, Kuechle M, Harlan JM, et al.: Fibroblast and endothelial apoptosis in systemic sclerosis. Curr Opin Rheumatol 2003, 15:756–760. 81 Parekh D, Ziegler W, Yonezawa K, et al.: Mammalian TOR controls one of two kinase pathways acting upon nPKCdelta and nPKCepsilon. J Biol Chem 1999, 274:34758–34764. Shegogue D, Trojanowska M: Mammalian target of rapamycin positively regulates collagen type I production via a phosphatidylinositol 3-kinaseindependent pathway. J Biol Chem 2004, 279:23166–23175. This work is the first direct demonstration of collagen gene regulation by mTOR. Because mTOR inhibitors (rapamycin) are clinically approved, this finding may have a potential therapeutic application. 59 •• 60 61 Kumar V, Pandey P, Sabatini D, et al.: Functional interaction between RAFT1/FRAP/mTOR and protein kinase cdelta in the regulation of capdependent initiation of translation. EMBO J 2000, 19:1087–1097. Leask A, Abraham DJ, Finlay DR, et al.: Dysregulation of transforming growth factor beta signaling in scleroderma: overexpression of endoglin in cutaneous scleroderma fibroblasts. Arthritis Rheum 2002, 46:1857–1865. 67 Yamakage A, Kikuchi K, Smith EA, et al.: Selective upregulation of plateletderived growth factor alpha receptors by transforming growth factor beta in scleroderma fibroblasts. J Exp Med 1992, 175:1227–1234. 68 Zhuo Y, Zhang J, Laboy M, et al.: Modulation of PDGF-C and PDGF-D expression during bleomycin-induced lung fibrosis. Am J Physiol Lung Cell Mol Physiol 2004, 286:L182–L188. 69 Ponten A, Li X, Thoren P, et al.: Transgenic overexpression of platelet-derived growth factor-C in the mouse heart induces cardiac fibrosis, hypertrophy, and dilated cardiomyopathy. Am J Pathol 2003, 163:673–682. 70 Klareskog L, Gustafsson R, Scheynius A, et al.: Increased expression of platelet-derived growth factor type B receptors in the skin of patients with systemic sclerosis. Arthritis Rheum 1990, 33:1534–1541. 71 Kawaguchi Y, Hara M, Wright TM: Endogenous IL-1alpha from systemic sclerosis fibroblasts induces IL-6 and PDGF-A. J Clin Invest 1999, 103:1253– 1260. Hu B, Wang S, Zhang Y, et al.: A nuclear target for interleukin-1alpha: interaction with the growth suppressor necdin modulates proliferation and collagen expression. Proc Natl Acad Sci USA 2003, 100:10008–10013. The findings of this study suggest a novel role for pre-IL-1␣ as an antagonist of antiproliferative and antifibrotic actions of necdin. 72 •• Animal models of systemic sclerosis: insights into systemic sclerosis pathogenesis and potential therapeutic approaches Paul J. Christner and Sergio A. Jimenez Purpose of review Animal models have been extremely valuable in contributing to a better understanding of the pathogenesis of systemic sclerosis. Discussed here are recent studies that have examined the molecular pathways and potential therapeutic approaches for systemic sclerosis using animal models. Recent findings Reported evidence further indicates that the immune system plays a role in modulating the fibrosis observed in the tight skin-1/+ mouse model for systemic sclerosis. CD19, interleukin-6, and interleukin-4 are involved. The injection of spleen cells into immune-compromised mice resulted in fibrotic, vascular, and immunologic alterations quite similar to those of systemic sclerosis. Transforming growth factor- and its signaling pathway (JAK kinase and STAT-6, Smad2/3, and Smad7) appear to play a central role in the development of fibrosis as well as monocyte chemoattractant protein-1, CCR-2, platelet-derived growth factor C, and excessive apoptosis. Viruses were shown to be possible cofactors. The therapeutic agents hepatocyte growth factor and halofuginone were shown to prevent fibrosis in animal models of systemic sclerosis. Summary The transforming growth factor- signaling pathway is a common mechanism of tissue fibrosis in animal models of systemic sclerosis, although numerous additional molecules modulate this pathway or have a direct effect on fibrosis. Keywords systemic sclerosis, animal models, Tsk (tight skin) mouse, bleomycin-induced scleroderma, transforming growth factor  Curr Opin Rheumatol 16:746–752. © 2004 Lippincott Williams & Wilkins. Division of Rheumatology, Department of Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA IL MAGP-2 MCP MMP PAI PDGF SSc TGF TIMP Tsk interleukin microfibril-associated glycoprotein 2 monocyte chemoattractant protein matrix metalloproteinase plasminogen activator inhibitor platelet-derived growth factor systemic sclerosis transforming growth factor tissue inhibitor of metalloproteinase tight skin © 2004 Lippincott Williams & Wilkins 1040–8711 Introduction Animal models of systemic sclerosis (SSc) have provided valuable insights into the causative mechanisms and the pathogenesis of SSc and have furnished the means to potentially test useful therapeutic interventions [1–3]. Among the numerous animal models for SSc, the most extensively studied are murine because of the large number of inbred mouse strains available and the detailed genetic and molecular information available for this species. Although the animal models described to date do not reproduce precisely all the clinical and pathologic alterations of the human disease, several of them show some of the typical abnormalities of this disorder, and prudent interpretation of the results obtained from their study has provided substantial and valuable information about the pathogenesis of SSc. Here we review recent studies using animal models of SSc, emphasizing their contribution to understanding the pathogenesis of the human disease and potential approaches for its treatment. Insights into pathogenesis and molecular pathways of systemic sclerosis Numerous recent studies have examined molecular pathways that may be of relevance to the pathogenesis of SSc. Supported by NIH grants AR32564 to S.A.J. and AR42666 to P.J.C. Correspondence to Sergio A. Jimenez, Thomas Jefferson University, Division of Rheumatology, 233 South 10th Street, Room 509 BLSB, Philadelphia, PA 19107, USA Tel: 215 503 5042; fax: 215 923 4649; e-mail: [email protected] Current Opinion in Rheumatology 2004, 16:746–752 Abbreviations BALF ECM HGF 746 bronchoalveolar lavage fluid extracellular matrix hepatocyte growth factor Studies with Tsk1 mice Saito et al. [4] reported that the level of the cytokine CD19 can influence skin fibrosis and autoimmunity in the tight skin (Tsk)1/+ mouse. Tsk1/+ mice were hyperresponsive to CD19 transmembrane signals and had a decreased expression of IgM expression, enhanced serum Ig levels, and spontaneous autoantibody production. The reason for this aberrant immune response appeared Animal models of systemic sclerosis Christner and Jimenez 747 to be the constitutive increase in tyrosine phosphorylation of CD19 in B cells from the Tsk1/+ mouse. In addition, CD19-mediated [Ca2+]i responses, Vav phosphorylation and Lyn kinase activity were increased. Furthermore, Tsk1/+ mice deficient in CD19 had significantly less skin fibrosis and showed a much higher expression of surface IgM on their B cells, did not develop the autoantibodies characteristic of Tsk1/+ animals, and had reduced interleukin (IL)-6 production. The authors concluded that chronic B cell activation resulting from augmented CD19 signaling could lead to skin sclerosis and autoantibody production through a mechanism of IL-6 overproduction. In another study, Kodera et al. [5] reported that the IL-4 gene was crucial for the expression of the Tsk1 phenotype. Tsk1/Tsk1 mice are not viable, and they die in utero at approximately the same time that expression of the mutated fibrillin 1 gene is initiated. These authors reported that elimination of either one copy or both copies of the IL-4 gene allowed survival of the homozygous Tsk1/Tsk1 mouse. Histopathology indicated that these mice did not show any cutaneous hyperplasia but still displayed pulmonary emphysema. In vitro experiments indicated that IL-4 regulated the levels of transforming growth factor (TGF)-, and the authors postulated that the downregulation of TGF- levels in these IL-4– deficient Tsk mice prevented the development of cutaneous fibrosis. The important conclusion of these studies is that IL-4 is crucial in the development of the Tsk phenotype and is involved in the mortality of Tsk/Tsk embryos. In a follow-up study, McGaha et al. [6] reported that IL-4 induced a substantial increase in collagen synthesis in both Tsk1/+ and normal mouse dermal fibroblasts, but the effect was greater in Tsk1/+ cells. They further showed that the IL-4 signaling cascade was altered in Tsk1/+ fibroblasts compared with control specimens. In Tsk1/+ cells, the phosphorylation of JAK kinases, which in turn phosphorylate the IL-4 receptor, was constitutive, whereas in cells from normal mice, IL-4 was required for JAK phosphorylation. Another signal transduction molecule, STAT-6, was also involved because IL-4 induced higher levels of phosphorylated STAT-6 in Tsk1/+ cells than in control cells. Transfection studies with a portion of the ␣2(I) collagen gene promoter showed that IL-4 and STAT-6 could upregulate its transcriptional activity and that AP-1 and Sp-1 transcription factors were involved in this effect. The authors concluded that type I collagen gene expression is enhanced by IL-4 either directly or through TGF-– mediated mechanisms. Another study characterized the tissue expression of elastic fiber–related proteins in Tsk1/+ mouse skin. Lemaire et al. [7] showed that Tsk mutant fibrillin 1 increases extracellular matrix (ECM) incorporation of microfibril-associated glycoprotein 2 (MAGP-2) and type I collagen. The authors analyzed the effect of the Fbn1 mutation present in the Tsk1/+ mouse on the structure and composition of the ECM by transfecting the mutated Fbn1 gene under the control of a tetracyclinedependent promoter into a mouse embryonic cell line. It was found that when the mutated Fbn1 was expressed in the mouse embryonic cell line, the sharply defined fibrillin network became smudged and blurred when examined by immunofluorescence. The fibrillin fibers produced in the presence of the mutated Fbn1 were also broader and less intensely stained. However, in contrast with previous studies, Col1a1 mRNA was equally expressed in the presence or absence of expression of the mutated Fbn1. This was true even when TGF-1 was added to the cultures. However, expression of the mutated Fbn1 had a significant effect on the deposition and incorporation of type I collagen molecules into the ECM. There was also an increase in the MAGP-2 fibrillar structures within the ECM caused by the expression of the mutated Fbn1. The authors also reported an increase in MAGP-2 deposits in lesional SSc skin and concluded that alterations in the microfibril structure or deposition may contribute to cutaneous fibrosis in SSc. Studies with the bleomycin-induced model of cutaneous fibrosis Murota et al. [8] examined the effects of disruption of the TNF-␣ receptor p55 on collagen turnover in skin fibroblasts from mice injected with bleomycin subcutaneously. The treatment caused mild sclerosis after 14 days in normal mice, whereas it resulted in severe sclerosis after only 3 days in TNF-␣ receptor p55−/− mutants. The mutant mice showed thickened and homogeneous collagen bundles and dermal inflammatory infiltrates, whereas at the same time-points the control mice had severe inflammatory infiltrates but no cutaneous sclerosis or fibrosis. After 3 days of bleomycin injections, the skin in the mutant mice was significantly thicker than that in the control mice, and the collagen content at the site of injection was almost threefold higher. At sites distant from the injection site, there was no increase in dermal thickness, and the expression levels of IL-4 and plateletderived growth factor (PDGF) were also not different. By contrast, TGF- mRNA was reduced and TNF-␣ mRNA was induced in a time-dependent and dosedependent manner in the mutant and control mice as a result of the bleomycin injections. Most interestingly, the expression level of Col1a1 mRNA was not altered in the mutant mice compared with control mice. However, bleomycin induced an increased expression of matrix metalloproteinase (MMP)-1 mRNA in the control mice, whereas MMP-1 expression was significantly less in the mutants. MMP-2, MMP-9, and gelatinase were equal in both sets of mice. These same results were confirmed when mouse embryonic fibroblasts were treated with bleomycin in vitro. The authors concluded that TNF-␣ receptor p55 is an essential component of the signaling 748 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes pathway for MMP-1 expression leading to the degradation of collagen. In another study, Takagawa et al. [9] examined the effects of repeated injections of bleomycin into the skin of mice, which induced inflammation and fibrosis by 1 week. They further characterized the role of cellular Smad3 expression in the skin using specific antibodies. Mice injected with bleomycin had abundant Smad3 throughout the dermis, epidermis, hair follicles, and sebaceous glands. This effect was absent in control mice injected with phosphate-buffered saline. At 3 days and 1 week, Smad3 was detected mainly in infiltrating macrophages; the inflammation began to resolve in 2 to 3 weeks, and Smad3 expression was confined mainly to the nuclei of fibroblasts. The authors also examined Smad 2 and Smad3 phosphorylation as an indicator of the activation level of the TGF-/Smad pathway. They showed that 70% of the lesional fibroblasts in mice injected with bleomycin were positive for phospho-Smad2/3 and that it was detectable even after 3 weeks of bleomycin injection, primarily in fibroblast nuclei. By contrast, phospho-Smad2/3 was undetectable in control mice. The authors concluded that activation of the Smad signaling pathway is ongoing in resident lesional fibroblasts even after the inflammation has resolved. In experiments with dermal fibroblasts, it was shown that bleomycin has no direct effect on the phosphorylation of Smad2/3 but that its effect is mediated by TGF-. To understand the mechanism by which the resident lesional fibroblasts continued to produce phospho-Smad2/3 in the absence of inflammation, the authors postulated that there might be a defect in Smad7 that downregulates TGF- activation of the Smad signaling pathway. In vivo expression of Smad7 in the dermis of TGF-–injected mice was demonstrated after 24 hours. By contrast, in the bleomycininjected mice Smad7 was induced in the infiltrating mononuclear cells but not in the fibroblasts. These results indicated that the ratio of phospho-Smad2/3 to Smad7-positive fibroblasts was significantly altered in bleomycin-injected mouse dermis and that this alteration was responsible for the cutaneous fibrosis that develops in these mice. In a related study, Yamamoto and Nishioka [10] examined the role of monocyte chemoattractant protein-1 (MCP-1) and its receptor, CCR-2, in the pathogenesis of bleomycin-induced scleroderma. The authors first examined the expression of MCP-1 and CCR-2 in the lesional skin of mice injected subcutaneously with bleomycin. They found that MCP-1 was weakly detectable on scattered mononuclear cells in control mice. In contrast, MCP-1 and CCR-2 were detectable on infiltrating mononuclear cells in the dermis of bleomycin-injected mice, and the number of positive cells peaked between 2 and 3 weeks during treatment. Positive fibroblasts were also detected in the sclerotic dermis at later stages. The au- thors concluded that there was concurrent upregulation of MCP-1 and CCR-2 in mononuclear cells at early inflammatory stages and in fibroblasts at a later sclerotic stage in the skin of bleomycin-injected mice. The authors next investigated the effect of antibodies against MCP-1 on the course of dermal fibrosis. Administration of the antibody every other day decreased the bleomycin-induced dermal fibrosis and reduced the number of infiltrating mononuclear cells compared with control animals injected with bleomycin but receiving no antibody. The link between MCP-1 and increased expression of Col1a1 mRNA was examined in cultured fibroblasts. The addition of MCP-1 to the fibroblast cultures increased collagen mRNA levels by more than fourfold. In addition, decorin mRNA levels were also increased. These mRNA increases were MCP-1 dose dependent. However, the levels of fibronectin and biglycan mRNA were not significantly altered. The authors concluded that MCP-1 may induce fibrosis by directly upregulating Col1a1 mRNA in fibroblasts as well as by exerting an indirect effect through cytokines released from immunocytes recruited into lesional skin. Another study by Yamamoto and Nishioka [11] examined the possible role of apoptosis in the pathogenesis of bleomycin-induced scleroderma. These authors showed that murine skin treated with bleomycin showed strong apoptotic signals indicative of cell death in the infiltrating mononuclear cells, hair follicles, and sebaceous glands in the dermis. The signals were present as early as 1 week and increased markedly after 3 weeks of bleomycin treatment. The molecular mechanism of the induced apoptosis involved Fas and FasL. Fas was detected in the cell membrane of some mononuclear cells after 1 week of bleomycin treatment and increased to a maximum after 2 weeks of treatment. Fas was also expressed constitutively in fibroblasts from 1 to 4 weeks of bleomycin treatment. FasL was detectable after 1 week of treatment in inflammatory cells and in fibroblastic cells at 3 to 4 weeks. Reverse transcription polymerase chain reaction showed that Fas mRNA was clearly detectable throughout all layers of skin during 1 to 4 weeks of bleomycin treatment, whereas FasL mRNA expression was upregulated and peaked at 7.5 times the control values at 3 weeks after the beginning of bleomycin treatment. The authors next investigated the role of caspase3, a downstream regulator of Fas/FasL. Immunohistochemistry showed that caspase-3 expression was detected in epidermis, hair follicles, and sebaceous glands but not in cellular infiltrates in control mice. In bleomycin-treated skin, caspase-3 was detected in both the nucleus and the cytoplasm of infiltrating mononuclear cells and a small portion of the fibroblasts. Reverse transcription polymerase chain reaction showed that both caspase-1 and caspase-3 mRNA expression was upregulated in lesional skin and peaked at 3 weeks of bleomycin treatment. The upregulation of caspase-3 fol- Animal models of systemic sclerosis Christner and Jimenez 749 lowed the same time course as FasL upregulation. An anti-FasL antibody partially prevented the development of dermal fibrosis after bleomycin treatment, and collagen content was reduced 50% compared with skin from controls treated with bleomycin and normal IgG. The number of apoptotic mononuclear cells in the sclerotic skin was also decreased. The authors conclude that there is a relation between the Fas/FasL system and caspase-3 activation that mediates apoptosis and that the continuous and extensive expression of FasL may participate in the development of cutaneous fibrosis/sclerosis by inducing excessive apoptosis or by modulating inflammatory mediators. Zhuo et al. [12] examined the modulation of PDGF-C and PDGF-D in bleomycin-induced pulmonary fibrosis in mice. PDGFs induce fibroblast proliferation and chemotaxis through cell surface receptor signaling. The authors reported that during bleomycin-induced lung fibrosis in mice, the levels of PDGF-C mRNA increased significantly, whereas the level of PDGF-D mRNA decreased. The increased levels of PDGF-C mRNA were localized by in situ hybridization to areas of lung fibrosis and were not observed in the lungs of bleomycin-resistant BALB/c mice. The authors suggested that PDGF-C is involved in the development of bleomycin-induced pulmonary fibrosis through binding to its receptor. Systemic sclerosis model induced by spleen cell injections in immunodeficient mice Ruzek et al. [13] described a modified model of induced SSc that demonstrates all major aspects of the human disease. The authors transferred donor B10.D2 spleen cells into RAG-2 knockout mice to induce a graft-versushost response. RAG-2 knockout mice are genetically deficient in mature T and B cells and therefore cannot generate an antigen-specific immune response. The results were similar to a graft-versus-host model using irradiated mice. In the treated mice, dermal thickening developed, primarily of the extremities, with less pronounced dermal thickening in dorsal or abdominal skin. The dermal thickening peaked at 3 to 5 weeks and began to decline by 6 weeks but did not completely disappear even by 22 weeks. Fibrosis of internal organs including the kidneys, the intestinal tract, and the liver was also observed, and, in contrast to the dermis, the visceral fibrosis continued to increase over time. Overexpression of both type VII and type III collagens in the skin and type III collagen in the kidneys was also observed. The authors also reported a significant decrease in the luminal ratio of blood vessels in both skin and kidney, indicating vasoconstriction and occlusion, which was progressive over the course of the disease. In addition, the expression of the potent vasoconstrictor ET-1 was increased. The smooth muscle marker ␣SMA showed that the cells surrounding vessels bearing this marker had morphologic changes that likely contributed to occlusion of the vessel lumen. The mice also showed a prominent early immune response up to 3 weeks, which resolved by 6 weeks. CD4+, CD8+ T cells and macrophages were shown to participate: they increased tenfold, sixfold, and eightfold, respectively, in the ear dermis. CD4+ and CD8+ cells also increased in the kidney. Of remarkable importance was the observation that ANAs with a speckled pattern developed in the mice and that Scl-70 antibodies were present in more than 90% of these mice. The authors concluded that this modified model of graft-versus-host—induced SSc shows all the major components of the human disease and can be used to test effective therapeutic agents. Models of pulmonary fibrosis Kuroki et al. [14] investigated the role of TNF-␣ in pulmonary inflammation and fibrosis induced by intratracheal injection of bleomycin. They used TNF-␣ knockout (TNF-␣−/−) mice in this study. They demonstrated that the number of inflammatory cells in the bronchoalveolar lavage fluid (BALF) peaked at 7 days in TNF␣+/+ mice and then decreased. By contrast, in TNF-␣−/− mice the number of inflammatory cells in BALF was persistently increased to 35 days after the instillation of bleomycin. The predominant cells present in the BALF of TNF-␣+/+ mice were macrophages, whereas in TNF␣−/− mice they were lymphocytes. When cells from lung tissue were taken 21 days after bleomycin instillation, the total cell and lymphocyte numbers were higher in the TNF-␣−/− mice than in the TNF-␣+/+ mice. Histologic examination of TNF-␣+/+ and TNF-␣−/− mouse lungs revealed lymphocytic and neutrophilic infiltration, thickening of the alveolar septa, and proliferation of fibroblasts in both mouse strains 14 days after bleomycin instillation. At day 21 no difference in hydroxyproline content of the lungs between either strain was observed. However, in the TNF-␣+/+ mice the inflammatory response gradually subsided, whereas in the TNF-␣−/− mice massive infiltration of lymphocytes persisted, and a fibrotic and honeycomb tissue morphology was observed 75 days after bleomycin instillation. TNF-␣ production was measured in the TNF-␣+/+ mice in response to bleomycin instillation and was shown to be biphasic reaching an initial peak after 12 hours, declining until 7 days and then rising again and remaining persistently elevated up to 50 days after instillation. Flow cytometry revealed that markers for the TNF␣ receptor were upregulated on inflammatory cells in BALF from TNF␣+/+ and TNF-␣−/− mice 14 days after bleomycin instillation. Further flow cytometric analysis revealed that significant numbers of the inflammatory cells in BALF from the TNF-␣+/+ mice were apoptotic, whereas fewer apoptotic cells were observed in the BALF from TNF␣−/− mice. When TNF-␣−/− mice were treated with murine rTNF-␣, the levels of apoptotic inflammatory cells increased, and two weekly treatments with the protein effectively diminished the pulmonary inflammatory response. The authors concluded that TNF-␣ is essential 750 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes for repressing pulmonary inflammation and fibrosis in bleomycin-induced pneumopathy and that this effect is mediated through induction of apoptosis of inflammatory cells. Potential therapeutic approaches A very interesting study examined the role of viruses as a cofactor in the development of pulmonary fibrosis in bleomycin-resistant mice. Lok et al. [15] studied the development of fibrosis in BALB/c mice injected intraperitoneally with bleomycin. This mouse strain is normally resistant to bleomycin-induced fibrosis. The mice received, in addition to the intraperitoneal injections of bleomycin, transnasal dosing with murine gammaherpes virus 68. Control mice received no virus and only phosphate-buffered saline injections. In mice that received both the virus and bleomycin, significantly more severe lung inflammation developed than in mice that received virus alone, bleomycin alone, or saline. In addition, the collagen content of the lung followed a similar pattern. The authors concluded that the virus does not cause pulmonary fibrosis by itself but can act as a cofactor in the development of lung fibrosis or in its progression. McGaha et al. [17] examined the effects of halofuginone, a drug with antifibrotic properties, in preventing the appearance of dermal fibrosis in the Tsk1/+ mouse. They reported that 1 µg of halofuginone injected intraperitoneally every other day for 60 days prevented the development of dermal fibrosis in both neonates and adult Tsk1/+ mice, as demonstrated by histologic analysis. The thickness of the skin of Tsk1/+ mice that had been treated with halofuginone was less than 70% of the untreated animal skin thickness. Dermal fibroblasts derived from these mice were also sensitive to the drug, which caused reduced type I collagen synthesis. The drug appeared to cause this effect by affecting the promoter activity of type I collagen genes. The site of action of halofuginone on the Col1a2 promoter was localized to a region between −3200 and +54 bp. The mechanism by which halofuginone exerted this inhibitory effect on collagen synthesis appeared to be through the TGF- signaling pathways by blocking the phosphorylation of Smad3. Studies in transgenic mice In a very interesting study, Denton et al. [16] examined TGF- signaling pathways involved in tissue fibrosis, using transgenic mice. The authors reported the development of transgenic mice that express a kinase-deficient human type II TGF- receptor (TRII⌬k) in fibroblasts. In previous work, TRII⌬k was shown to act as a dominant negative inhibitor of TGF- signaling. However, in the present study, the authors demonstrated that in adult mice that expressed this receptor, pulmonary and dermal fibrosis developed. Neonatal dermal fibroblasts cultured from the transgenic mice proliferated more rapidly than control cells and produced more ECM. Several markers of TGF- signaling were upregulated, including plasmogen activator inhibitor-1, CTGF, and Smads 3, 4, and 7. Microarray experiments showed that the transgenic fibroblasts had a similar gene expression profile to that of littermate control fibroblasts that had been treated with TGF-1. TGF- was not able to further stimulate the transgenic fibroblasts. However, overexpression of type II TGF- receptors partially restored the responsiveness of these transgenic fibroblasts to TGF- stimulation. Additional studies of the mitogenactivated protein kinase pathways (another TGF- signaling pathway separate from the Smad signaling pathway) showed that they were less perturbed by recombinant TGF-, and the authors concluded that this pathway is less affected in the transgenic mice than the Smad pathways. Therefore, this mouse appears to be a model for the study of TGF- overexpression and signaling and the molecular pathways leading to tissue fibrosis. Animal models have also been used in numerous studies to examine putative therapeutic interventions that may be effective to modulate or improve the pathologic alterations typically present in patients with SSc. In a parallel paper, McGaha et al. [18] examined the effect of halofuginone on the development of the Tsk1/+ phenotype. The authors administered halofuginone intraperitoneally to newborn (1-day-old) and 1-month-old Tsk1/+ mice every other day for 60 days. They confirmed their earlier work, which showed that this drug significantly reduces collagenous material in Sirus-red stained skin sections. The collagen content of skin was decreased by 20 to 25% when drug treatment was begun in 1-month-old mice and by almost 50% when neonates were treated. In situ hybridization studies indicated that halofuginone-treated neonate mice had a significantly lower number of cells expressing type I collagen mRNA and that this number was indistinguishable from the number detected in C57BL/6 pa/pa normal control mice. The authors concluded that the decreased collagen content observed in halofuginone-treated Tsk1/+ mice is due to decreased numbers of cells expressing collagen in the dermis. The authors also presented data showing that halofuginone had no effect on the emphysema that develops in Tsk1/+ mice. An interesting observation was that when adult Tsk1/+ mice were treated with the drug, the level of anti–topoisomerase-1 and anti–fibrillin-1 antibodies typically present in these animals decreased to control levels in adult Tsk1/+ mice but not in neonates. A third study by McGaha et al. [19] examined the mechanisms of the inhibitory effects of halofuginone on fibrosis. They showed that the inhibition of collagen gene promoter activity caused by the drug is mediated Animal models of systemic sclerosis Christner and Jimenez 751 through transcription factors that regulate type I collagen gene expression. Using mouse-cultured fibroblasts, the authors found that TGF-1, PDGF, and PMA induced rapid phosphorylation of c-Jun, a component of a dimeric transcription factor that binds with highest affinity to the AP-1 regulatory element. In the presence of halofuginone, the induced phosphorylation of c-Jun was higher than in its absence, indicating that the mechanism that suppresses collagen synthesis may involve c-Jun. Further experiments showed that TGF-1–induced AP-1 binding activity was greatly increased in intensity, and the time over which the increase occurred was lengthened in the presence of halofuginone. Transfection assays also showed that there was a synergistic effect of TGF-1 and halofuginone on the activation of AP-1 activity and that this AP-1 complex induced by TGF-1 in the presence of halofuginone is a potential antagonist of type I collagen gene activity. Halofuginone abrogated the TGF-1–induced upregulation of the Col1a2 promoter and the reduction of Col1a2 mRNA levels through a c-Jun dependent mechanism. In additional tests on Tsk1/+ mice, the authors showed that the topical application of halofuginone led to increased amounts of phospho-c-Jun in the stratum and granular basal layer of the epidermis. Another potential therapeutic agent was studied by Gong et al. [20]. The authors showed that the recently described hepatocyte growth factor (HGF) modulates matrix metalloproteinases and plasminogen activator/plasmin proteolytic pathways in progressive renal interstitial fibrosis. HGF increased collagen catabolism in human proximal tubular epithelial cells (HKC) treated with TGF-1 associated with increased MMP activity and plasminogen activator/plasmin proteolytic pathway enhancement. HGF also induced the production of MMP-9 and prevented TGF-1–induced production of tissue inhibitor of metalloproteinase (TIMP)-2 and plasminogen activator inhibitor (PAI)-1. Continuous infusion of HGF in the rat remnant kidney model ameliorated renal fibrosis and tubulo-interstitial collagen deposition. In this model the authors also reported increased tubular expression of MMP-9, enhanced in situ gelatinolytic activity, restoration of plasmin activity, and decreased expression of TIMP-2 and PAI-1. There was an overall increase in TIMP-3 expression. Anti-HGF antibodies caused increased renal fibrosis and exaggerated accumulation of interstitial collagen deposition. Accompanying these changes were decreased tubular expression of MMP-9 and elevated TIMP-2 and PAI-1 expression. The authors concluded that HGF ameliorates renal fibrosis by enhancing ECM catabolism through the MMP and PA/plasmin proteolytic pathways. An additional study examined the possibility that HGF may have antifibrotic effects. Wu et al. [21] showed that HGF both prevents and ameliorates bleomycin-induced dermal sclerosis. These authors transfected a construct encoding human HGF into mouse skeletal muscle and showed that expression of HGF had a marked effect on preventing dermal sclerosis in mice injected with bleomycin. The HGF was effective even when injected 4 weeks after bleomycin treatment. Levels of HGF mRNA and protein were higher in skin, lung, muscle, and serum after two transfections. It appears that the effect of HGF in this model involves TGF-, because levels of TGF- were reduced. Transfections of HGF also ameliorated lung fibrosis. The authors suggest that gene therapy with HGF may be useful to prevent even established tissue fibrosis. Conclusion Most of the work published over the past 2 years indicates that the TGF- signaling pathway is a common mechanism by which fibrosis occurs. However, there are numerous other molecules that can either modulate this pathway or have a direct effect on fibrosis. Two compounds, HGF and halofuginone, have been shown to be effective in preventing or ameliorating fibrosis, and it appears that perturbation of TGF- pathways was involved in the mechanism of action of these drugs. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • Of special interest •• Of outstanding interest 1 Zhang JC, Gilliam AC: Animal models for scleroderma: an update. Curr Rheumatol Rep 2002, 4:150–162. 2 Jimenez SA, Christner PJ: Murine animal models of systemic sclerosis. Curr Opin Rheumatol 2002, 14:671–680. 3 Christner PJ, Jimenez SA: Spontaneous mouse models of systemic sclerosis. In Animal Models of Human Inflammatory Skin Diseases. Edited by Chan LS. Boca Raton, FL: CRC Press; 2004:495–515. 4 Saito E, Fujimoto M, Hasegawa M, et al.: CD19-dependent B lymphocyte signaling thresholds influence skin fibrosis and autoimmunity in the tight-skin mouse. J Clin Invest 2002, 109:1453–1462. 5 Kodera T, McGaha TL, Phelps R, et al.: Disrupting the IL-4 gene rescues mice homozygous for the tight-skin mutation from embryonic death and diminishes TGF production by fibroblasts. Proc Natl Acad Sci USA 2002, 99:3800– 3805. 6 McGaha TL, Le M, Kodera T, et al.: Molecular mechanisms of interleukin-4induced up-regulation of type I collagen gene expression in murine fibroblasts. Arthritis Rheum 2003, 48:2275–2284. 7 Lemaire R, Farina G, Kissin E, et al.: Mutant fibrillin 1 from tight skin mice increases extracellular matrix incorporation of microfibril-associated glycoprotein 2 and type I collagen. Arthritis Rheum 2004, 50:915–926. 8 Murota H, Hamasaki Y, Nakashima T, et al.: Disruption of tumor necrosis factor receptor p55 impairs collagen turnover in experimentally induced sclerodermic skin fibroblasts. Arthritis Rheum 2003, 48:1117–1125. 9 Takagawa S, Lakos G, Mori Y, et al.: Sustained activation of fibroblast transforming growth factor-/Smad signaling in a murine model of scleroderma. J Invest Dermatol 2003, 12:41–50. 10 Yamamoto T, Nishioka K: Role of monocyte chemoattractant protein-1 and its receptor, CCR-2, in the pathogenesis of bleomycin-induced scleroderma. J Invest Dermatol 2003, 12:510–516. 11 Yamamoto T, Nishioka K: Possible role of apoptosis in the pathogenesis of bleomycin-induced scleroderma. J Invest Dermatol 2004, 122:44–50. 752 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes 12 Zhuo Y, Zhang J, Laboy M, et al.: Modulation of PDGF-C and PDGF-D expression during bleomycin-induced lung fibrosis. Am J Physiol Lung Cell Mol Physiol 2004, 286:L182–L188. 13 Ruzek MC, Jha S, Ledbetter S, et al.: A modified model of graft-versus-hostinduced systemic sclerosis (scleroderma) exhibits all major aspects of the human disease. Arthritis Rheum 2004, 50:1319–1331. 14 Kuroki M, Noguchi Y, Shimono M, et al.: Repression of bleomycin-induced pneumopathy by TNF. J Immunol 2003, 170:567–574. 15 Lok SS, Haider Y, Howell D, et al.: Murine gammaherpes virus as a cofactor in the development of pulmonary fibrosis in bleomycin resistant mice. Eur Respir J 2002, 20:1228–1232. 16 Denton CP, Zheng B, Evans LA, et al.: Fibroblast-specific expression of a kinase-deficient type II transforming growth factor  (TGF) receptor leads to paradoxical activation of TGF signaling pathways with fibrosis in transgenic mice. J Biol Chem 2003, 278:25109–25119. 17 McGaha TL, Phelps RG, Spiera H, et al.: Halofuginone, an inhibitor of type-I collagen synthesis and skin sclerosis, blocks transforming-growth-factor-mediated Smad3 activation in fibroblasts. J Invest Dermatol 2002, 118:461– 470. 18 McGaha T, Kodera T, Phelps R, et al.: Effect of halofuginone on the development of tight skin (TSK) syndrome. Autoimmunity 2002, 35:277–282. 19 McGaha TL, Kodera T, Spiera H, et al.: Halofuginone inhibition of COL1A2 promoter activity via a c-Jun-dependent mechanism. Arthritis Rheum 2002, 46:2748–2761. 20 Gong R, Rifai A, Tolbert EM, et al.: Hepatocyte growth factor modulates matrix metalloproteinases and plasminogen activator/plasmin proteolytic pathways in progressive renal interstitial fibrosis. J Am Soc Nephrol 2003, 14: 3047–3060. 21 Wu M-H, Yokozeki H, Takagawa S, et al.: Hepatocyte growth factor both prevents and ameliorates the symptoms of dermal sclerosis in a mouse model of scleroderma. Gene Ther 2004, 11:170–180. Erratum In the September 2004 issue of Current Opinion in Rheumatology, a figure printed incorrectly. Figure 1 in “Apoptosis and estrogen deficiency in primary Sjögren syndrome” (Hayashi Y, Arakaki R, and Ishimaru N, Curr Opin Rheumatol 16:522–526) is reprinted below. The author apologizes for this error. Figure 1. An organ-specific autoantigen may play an important role on down-modulation of AICD A cleavage product of 120-kD ␣-fodrin in the target cells could be induced by estrogen deficiency during apoptosis through caspase activation, in particular caspase 1. Activation-induced cell death (AICD) results from the interaction between Fas and FasL, and activated T cells expressing both Fas and FasL are usually killed either by themselves or by interacting with each other. FasL undergo matrix metalloproteinase (MMP)-mediated proteolytic processing in their extracellular domains, resulting in the release of soluble FasL (sFasL). FasL-mediated AICD is down-regulated by autoantigen stimulation, indicating that the increased generation of soluble FasL inhibits the normal AICD process, leading to the proliferation of autoreactive CD4+ T cell. A defect in AICD may result in the development of autoimmune diseases. AICD, activation-induced cell death; MMP, matrix metalloproteinase; sFasL, soluble FasL. 753 Bibliography Current World Literature This bibliography is compiled by clinicians from the journals listed at the end of this publication. It is based on literature entered into our database between July 1, 2003 and June 30, 2004 (articles are generally added to the database about two and a half months after publication). In addition, the bibliography contains every paper annotated by reviewers; these references were obtained from a variety of bibliographic databases and published between the beginning of the review period and the time of going to press. The bibliography has been grouped into topics that relate to the reviews in this issue. Contents • Papers considered by the reviewers to be of special interest. •• Papers considered by the reviewers to be of outstanding interest. The number in square brackets following a selected paper, for example [7], refers to its number in the annotated references of the corresponding review. Current Opinion in Rheumatology 2004, 16:754–778 © 2004 Lippincott Williams & Wilkins ISSN 1040–8711 Myositis and myopathies Clinical assessment in adults Review (pp 668–672) Akesson A, Fiori G, Krieg T, et al.: Assessment of skin, joint, tendon and muscle involvement. Clin Exp Rheumatol 2003, 21:S5–S8. Bohlega S, Lach B, Meyer BF, et al.: Autosomal dominant hyaline body myopathy - Clinical variability and pathologic findings. Neurology 2003, 61:1519–1523. Botsios C, Ostuni P, BoscoloRizzo P, et al.: Dermatomyositis and malignancy of the pharynx in Caucasian patients: report of two observations. Rheumatol Int 2003, 23:309–311. Burns TM, Jones HR, Phillips LH, et al.: Clinically disparate stiff-person syndrome with GAD65 autoantibody in a father and daughter. Neurology 2003, 61:1291–1293. Carter JD, Valeriano J, Vasey FB: Aldolase levels in dermatomyositis and polymyositis with normal creatine kinase levels - Dr. Carter, et al reply. J Rheumatol 2003, 30:2078. Dalakas MC, Hohlfeld R: Diagnostic criteria for polymyositis and dermatomyositis - Reply. Lancet 2003, 362:1763. Dalakas MC, Hohlfeld R: Polymyositis and dermatomyositis. Lancet 2003, 362:971–982. Elst EF, Kamphuis SSM, Prakken BJ, et al.: Severe central nervous system involvement in juvenile dermatomyositis. J Rheumatol 2003, 30:2059–2063. 754 Vol 16 No 6 November 2004 768 Autoantibodies: pathogenetic significance, clinical utility 769 Myofibroblasts, EMT, cellular plasticity in tissue repair, fibrosis: potential implications for scleroderma Myositis and myopathies 754 Clinical assessment in adults 754 Clinical assessment in children 755 The use of imaging to assess patients with muscle disease 769 Recent advances in fibroblast signaling and biology in scleroderma 755 Is it really myositis? A consideration of the differential diagnosis 770 Animal models: insights into pathogenesis, genetic risk factors, potential therapeutic approaches 756 Myositis and autoantibodies 770 756 An update on pathogenesis 757 The immunogenetics of myositis Recent advances in the pathophysiology, diagnosis, staging, and management of pulmonary hypertension in scleroderma/SSc 757 New ideas about treatment 770 Anti-TGF-beta therapy in fibrosis and scleroderma 757 Miscellaneous 770 Treatment Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes 771 Renal fibrosis 771 Hepatic fibrosis 767 771 SLE 771 Miscellaneous Raynaud and vascular manifestations: pathophysiology, diagnosis, and management Isenberg DA, Allen E, Farewell V, et al.: International consensus on outcome measures for patients with idiopathic inflammatory myopathies. Development and initial validation of myositis activity and damage indices in patients with adult onset disease. Rheumatology 2003, 42:1–7. Isenberg DA, Allen E, Farewell V, et al.: International consensus outcome measures for patients with idiopathic inflammatory myopathies. Development and initial validation of myositis activity and damage indices in patients with adult onset disease. Rheumatology 2004, 43:49–54. Isenberg DA, Allen E, Farewell V, et al.: International consensus outcomes measures for patients • with idiopathic inflammatory myopathies. Development and initial validation of myositis activity and damage indices in patients with adult onset disease. Rheumatology 2004, 43:49– 54. [19]. Liozon E, Vidal E: Aldolase levels in dermatomyositis and polymyositis with normal creatine kinase levels. J Rheumatol 2003, 30:2077–2078. Marie I, Fournet P, Janvresse A, et al.: Periarticular calcifications and arthropathy as the first manifestation of polymyositis. Clin Exp Rheumatol 2003, 21:681–682. Mercado U: Aldolase levels in dermatomyositis and polymyositis with normal creatine kinase levels - Dr. Mercado replies. J Rheumatol 2003, 30:2078. Miller FW, Rider LG, Plotz PH, et al.: Diagnostic criteria for polymyositis and dermatomyositis. Lancet 2003, 362:1762–1763. Mok CC, Tsui EYK, Chau SY: Erosive arthropathy in amyopathic dermatomyositis. Clin Exp Rheumatol 2003, 21:409–410. Oddis CV: Idiopathic inflammatory myopathy - Management and prognosis. Rheum Dis Clin North Am 2002, 28:979–1001. Rider LG, Giannini EH, Harris-Love M, et al.: Defin•• ing clinical improvement in adult and juvenile myositis. J Rheumatol 2003, 30:603–617. [3]. Rider LG: Outcome assessment in the adult and juvenile idiopathic inflammatory myopathies. Rheum Dis Clin North Am 2002, 28:935–977. Silva CA, Sultan SM, Isenberg DA: Pregnancy outcome in adult-onset idiopathic inflammatory myopathy. Rheumatology 2003, 42:1168– 1172. Taivassalo T, Jensen TD, Kennaway N, et al.: The spectrum of exercise tolerance in mitochondrial myopathies - A study of 40 patients. Brain 2003, 126:413–423. vanderMeulen MFG, Bronner IM, Hoogendijk JE, et al.: Polymyositis - An overdiagnosed entity. Neurology 2003, 61:316–321. Clinical assessment in children Review (pp 673–677) Akesson A, Fiori G, Krieg T, et al.: Assessment of skin, joint, tendon and muscle involvement. Clin Exp Rheumatol 2003, 21:S5–S8. Burns TM, Jones HR, Phillips LH, et al.: Clinically disparate stiff-person syndrome with GAD65 autoantibody in a father and daughter. Neurology 2003, 61:1291–1293. Carter JD, Valeriano J, Vasey FB: Aldolase levels in dermatomyositis and polymyositis with normal Myositis and myopathies: The use of imaging to assess patients with muscle disease 755 creatine kinase levels - Dr. Carter, et al reply. J Rheumatol 2003, 30:2078. Dalakas MC, Hohlfeld R: Diagnostic criteria for polymyositis and dermatomyositis - Reply. Lancet 2003, 362:1763. Dalakas MC, Hohlfeld R: Polymyositis and dermatomyositis. Lancet 2003, 362:971–982. Hedin H, Larsson S: Muscle strength in children treated for displaced femoral fractures by external fixation - 31 patients compared with 31 matched controls. Acta Orthop Scand 2003, 74:305–311. Hicks JE, Drinkard B, Summers RM, et al.: Decreased aerobic capacity in children with juvenile dermatomyositis. Arthritis Rheum Arthritis Care Res 2002, 47:118–123. Huber AM, Feldman BM, Rennebohm RM, et al.: Validation and clinical significance of the childhood myositis assessment scale for assessment of muscle function in the juvenile idiopathic inflammatory myopathies. Arthritis Rheum 2004, 50:1595–1603. Liozon E, Vidal E: Aldolase levels in dermatomyositis and polymyositis with normal creatine kinase levels. J Rheumatol 2003, 30:2077–2078. Mendez EP, Lipton R, RamseyGoldman R, et al.: US incidence of juvenile dermatomyositis, 1995-1998: Results from the National Institute of Arthritis and Musculoskeletal and Skin Diseases Registry. Arthritis Rheum Arthritis Care Res 2003, 49:300–305. Mercado U: Aldolase levels in dermatomyositis and polymyositis with normal creatine kinase levels - Dr. Mercado replies. J Rheumatol 2003, 30:2078. Miller FW, Rider LG, Plotz PH, et al.: Diagnostic criteria for polymyositis and dermatomyositis. Lancet 2003, 362:1762–1763. Oddis CV: Idiopathic inflammatory myopathy - Management and prognosis. Rheum Dis Clin North Am 2002, 28:979–1001. Pachman LM: Juvenile dermatomyositis - Immunogenetics, pathophysiology, and disease expression. Rheum Dis Clin North Am 2002, 28:579–592. Pachman LM: Juvenile dermatomyositis: immunogenetics, pathophysiology, and disease expression. Rheum Dis Clin North Am 2002, 28:579–602. Rider LG, Giannini EH, Harris-Love M, et al.: Defining clinical improvement in adult and juvenile myositis. J Rheumatol 2003, 30:603–617. Rider LG: Outcome assessment in the adult and juvenile idiopathic inflammatory myopathies. Rheum Dis Clin North Am 2002, 28:935–977. Ruff C: Growth in bone strength, body size, and muscle size in a juvenile longitudinal sample. Bone 2003, 33:317–329. Ruperto N, Ravelli A, Murray KJ, et al.: Preliminary core sets of measures for disease activity and damage assessment in juvenile systemic lupus erythematosus and juvenile dermatomyositis. Rheumatology 2003, 42:1452–1459. Siegel KL, Hicks JE, Koziol DE, et al.: Walking ability and its relationship to lower-extremity muscle strength in children with idiopathic inflammatory myopathies. Arch Phys Med Rehabil 2004, 85:767–771. Stathopulu E, Baildam E: Gower’s sign in children with juvenile idiopathic arthritis. Rheumatology 2003, 42:1002–1003. Stewart WA, Acott PD, Salisbury SR, et al.: Bone mineral density in juvenile dermatomyositis Assessment using dual x-ray absorptiometry. Arthritis Rheum 2003, 48:2294–2298. vanderMeulen MFG, Bronner IM, Hoogendijk JE, et al.: Polymyositis - An overdiagnosed entity. Neurology 2003, 61:316–321. The use of imaging to assess patients with muscle disease Review (pp 678–683) Asakawa DS, Nayak KS, Blemker SS, et al.: Realtime imaging of skeletal muscle velocity. J Magn Reson Imaging 2003, 18:734–739. Ayyar R: MR imaging of the lower leg versus clinical electrophysiologic examination in the differential diagnosis of neurogenic foot drop. AJNR Am J Neuroradiol 2003, 24:1273–1274. Hengstman GJD: Magnetic resonance imaging criteria to differentiate inclusion body myositis from polymyositis. J Rheumatol 2003, 30:1892. Kane D, Balint PV, Wood F, et al.: Early diagnosis of pyomyositis using clinic-based ultrasonography in a patient receiving infliximab therapy for Behcet’s disease. Rheumatology 2003, 42: 1564–1565. Kane D, Grassi W, Sturrock R, et al.: Musculoskeletal ultrasound - A state of the art review in • rheumatology. Part 2. Clinical indications for musculoskeletal ultrasound in rheumatology. Rheumatology (Oxford) 2004, 43:829–838. [17]. Kapur S, Brunet JA, McKendry RJ: Diabetic muscle infarction - Case report and review. J Rheuma• tol 2004, 31:190–194. [24]. Lehtinen J, Tingart MJ, Apreleva M, et al.: Practical assessment of rotator cuff muscle volumes using shoulder MRI. Acta Orthop Scand 2003, 74:722–729. Maillard SM, Jones R, Owens C, et al.: Quantitative assessment of MRI T-2 relaxation time of thigh muscles in juvenile dermatomyositis. Rheumatology 2004, 43:603–608. Maillard SM, Jones R, Owens C, et al.: Quantitative •• assessment of MRI T2 relaxation time of thigh muscles in juvenile dermatomyositis. Rheumatology (Oxford) 2004, 43:603–608. [39]. Pfleiderer B, Lange J, Loske KD, et al.: Metabolic •• disturbances during short exercises in dermatomyositis revealed by real-time functional 31P magnetic resonance spectroscopy. Rheumatology (Oxford) 2004, 43:696–703. [45]. Trusen A, Beissert M, Schultz G, et al.: Ultrasound and MRI features of pyomyositis in children. • Eur Radiol 2003, 13:1050–1055. [19]. Verrall GM, Slavotinek JP, Barnes PG, et al.: Diagnostic and prognostic value of clinical findings in 83 athletes with posterior thigh injury Comparison of clinical findings with magnetic resonance Imaging documentation of hamstring muscle strain. Am J Sports Med 2003, 31:969–973. Yanagisawa O, Niitsu M, Yoshioka H, et al.: MRI determination of muscle recruitment variations in dynamic ankle plantar flexion exercise. Am J Phys Med Rehabil 2003, 82:760–765. Is it really myositis? A consideration of the differential diagnosis Review (pp 684–691) Allen E, Schmahmann S, Sauser D, et al.: Fasciitis in amyopathic dermatomyositis. JCR - J Clin Rheumatol 2003, 9:51–53. Askanas V, Engel WK: Proposed pathogenetic cascade of inclusion-body myositis - Importance • of amyloid-beta, misfolded proteins, predisposing genes, and aging. Curr Opin Rheumatol 2003, 15:737–744. [3]. Ballantyne CM, Corsini A, Davidson MH, et al.: Risk for myopathy with statin therapy in high risk patients. Arch Intern Med 2003, 163:553– 564. Bansal D, Miyake K, Vogel SS, et al.: Defective •• membrane repair in dysferlin-deficient muscular dystrophy. Nature 2003, 423:168–172. [32]. Bassez G, Authier FJ, LechaptZalcman E, et al.: Inflammatory myopathy with abundant macro- phages (IMAM): A condition sharing similarities with cytophagic histiocytic panniculitis and distinct from macrophagic myofasciitis. J Neuropathol Exp Neurol 2003, 62:464–474. Better OS, Rubinstein I, Reis DN: Muscle crush compartment syndrome: fulminant local edema with threatening systemic effects. Kidney Int 2003, 63:1155–1157. Botsios C, Ostuni P, BoscoloRizzo P, et al.: Dermatomyositis and malignancy of the pharynx in Caucasian patients: report of two observations. Rheumatol Int 2003, 23:309–311. Caporali R, Cavagna L, Bellosta M, et al.: Inflammatory myopathy in a patient with cutaneous findings of pityriasis rubra pilaris: a case of Wong’s dermatomyositis. Clin Rheumatol 2004, 23:63–65. Christopoulos C, Savva S, Pylarinou S, et al.: Localised gastrocnemius myositis in Crohn’s disease. Clin Rheumatol 2003, 22:143–145. Confalonieri P, Oliva L, Andreetta F, et al.: Muscle inflammation and MHC class I up-regulation in • muscular dystrophy with lack of dysferlin - An immunopatholgical study. J Neuroimmunol 2003, 142:130–136. [25]. Dalakas MC, Hohlfeld R: Diagnostic criteria for polymyositis and dermatomyositis - Reply. Lancet 2003, 362:1763. Derk CT, Vivino FB, Kenyon L, et al.: Inclusion body myositis in connective tissue disorders: case report and review of the literature. Clin Rheumatol 2003, 22:324–328. Eklund KK, Anttila P, LeirisaloRepo M: Eosinophilic fasciitis, myositis and arthritis as early manifestations of peripheral T-cell lymphoma. Scand J Rheumatol 2003, 32:379–380. Guis S, Mattei JP, Liote F: Drug-induced and toxic myopathies. Best Pract Res Clin Rheumatol 2003, 17:877–907. Hinderlich S, Salama I, Eisenberg I, et al.: Distal myopathy with rimmed vacuoles is allelic to hereditary inclusion body myopathy. Neurology 2003, 61:145. Horvath R, Lochmuller H, Scharfe C, et al.: A tRNA(Ala) mutation causing mitochondrial myopathy clinically resembling myotonic dystrophy. J Med Genet 2003, 40:752–757. Hyman MH: Issues in statin-associated myopathy. JAMA 2003, 290:888. Hyman MH: Statin-associated myopathy with normal creatine kinase levels. Ann Intern Med 2003, 138:1007. Juarez M, Misischia R, Alarcon GS: Infections in systemic connective tissue diseases: systemic lupus erythematosus, scleroderma, and polymyositis/dermatomyositis. Rheum Dis Clin North Am 2003, 29:163–184. Kaneda D, Sugie K, Yamamoto A, et al.: A novel form of autophagic vacuolar myopathy with late-onset and multiorgan involvement. Neurology 2003, 61:128–131. King RJ, Laugharne D, Kerslake RW, et al.: Primary obturator pyomyositis: A diagnostic challenge. J Bone Joint Surg (Br) 2003, 85B:895–898. Kirschner J, Bonnemann CG: The congenital and limb-girdle muscular dystrophies - Sharpening • the focus, blurring the boundaries. Arch Neurol 2004, 61:189–199. [28]. Kissin EY, Corbo JC, Farraye FA, et al.: Colchicine myopathy in a patient with familial Mediterranean fever and normal renal function. Arthritis Rheum Arthritis Care Res 2003, 49:614–616. Luh JY, Karnath BM: Issues in statin-associated myopathy. JAMA 2003, 290:888. Maeshima E, Nishimoto T, Yamashita M, et al.: Progressive systemic sclerosis-polymyositis overlap syndrome with eosinophilic pleural effusion. Rheumatol Int 2003, 23:252–254. Massawi G, Hickling P, Hilton D, et al.: Inclusion body myositis evolving in systemic lupus erythrematosus? A case report. Rheumatology 2003, 42:U9–1014. Mastaglia FL, Garlepp MJ, Phillips BA, et al.: Inflam•• matory myopathies - Clinical, diagnostic and 756 Myositis and myopathies: Myositis and autoantibodies therapeutic aspects. Muscle Nerve 2003, 27: 407–425. [6]. Miller FW, Rider LG, Plotz PH, et al.: Diagnostic criteria for polymyositis and dermatomyositis. Lancet 2003, 362:1762–1763. Muntoni F, Torelli S, Ferlini A: Dystrophin and muta•• tions - One gene, several proteins, multiple phenotypes. Lancet Neurol 2003, 2:731–740. [40]. Musial J, Undas A, CelinskaLowenhoff M: Polymyositis associated with infliximab treatment for rheumatoid arthritis. Rheumatology 2003, 42: 1566–1568. Narvaez J, VilasecaMomplet J: Severe acute myopathy induced by minocycline. Am J Med 2004, 116:282–283. Nishino I, Noguchi S: Distal myopathy with rimmed vacuoles is allelic to hereditary inclusion body myopathy - Reply. Neurology 2003, 61:145. Phillips PS, Haas RH: Statin-associated myopathy with normal creatine kinase levels - In response. Ann Intern Med 2003, 138:1008– 1009. Richter JG, Becker A, Ostendorf B, et al.: Differential diagnosis of high serum creatine kinase levels in systemic lupus erythematosus. Rheumatol Int 2003, 23:319–323. Selcen D, Ohno K, Engel AG: Myofibrillar myopathy: clinical, morphological and genetic studies in 63 patients. Brain 2004, 127:439–451. Suehiro S, Shiokawa S, Taniguchi S, et al.: Gallium67 scintigraphy in the diagnosis and management of chronic sarcoid myopathy. Clin Rheumatol 2003, 22:146–148. Taylor RW, Schaefer AM, Barron MJ, et al.: The diagnosis of mitochondrial muscle disease. • Neuromuscul Disord 2004, 14:237–245. [53]. Teichholz LE: Statin-associated myopathy with normal creatine kinase levels. Ann Intern Med 2003, 138:1008. Thompson PD, Clarkson P, Karas RH: Issues in statin-associated myopathy - Reply. JAMA 2003, 290:888–889. Toma E, Loignon M: Statin-associated myopathy with normal creatine kinase levels. Ann Intern Med 2003, 138:1007–1008. Torgovnick J, Arsura E: Statin-associated myopathy with normal creatine kinase levels. Ann Intern Med 2003, 138:1007. Tsuruta Y, Ikezoe K, Nakagaki H, et al.: A case of dermato-fasciitis: amyopathic dermatomyositis associated with fasciitis. Clin Rheumatol 2004, 23:160–162. Wagner B, KaganHallet KS, Russell IJ: Concomitant presentation of adermatopathic dermatomyositis, statin myopathy, fibromyalgia syndrome, piriformis muscle myofascial pain syndrome, and diabetic neuropathy. J Musculoskeletal Pain 2003, 11:25–30. Wang JY, Lee LN, Hsueh PR, et al.: Tuberculous myositis: a rare but existing clinical entity. Rheumatology 2003, 42:836–840. Wessig C, Klein R, Schneider MF, et al.: Neuropathology and binding studies in anti-amphiphysin-associated stiff-person syndrome. Neurology 2003, 61:195–198. Yabe I, Higashi T, Kikuchi S, et al.: GNE mutations causing distal myopathy with rimmed vacuoles • with inflammation. Neurology 2003, 61:384– 386. [57]. Zatz M, dePaula F, Starling A, et al.: The 10 autoso•• mal recessive limb-girdle muscular dystrophies. Neuromuscul Disord 2003, 13:532– 544. [29]. vanderMeulen MFG, Bronner IM, Hoogendijk JE, et •• al.: Polymyositis - An overdiagnosed entity. Neurology 2003, 61:316–321. [7]. Myositis and autoantibodies Review (pp 692–699) Howard OMZ, Dong HJF, Yang D, et al.: Histidyl•• tRNA synthetase and asparaginyl-tRNA synthetase, autoantigens in myositis, activate che- mokine receptors on T lymphocytes and immature dendritic cells. J Exp Med 2002, 196:781–791. [41]. Kamei H: Intracellular localization of histidyl-tRNA synthetase/Jo-1 antigen in T24 cells and some • other cells. J Autoimmun 2004, 22:201–210. [35]. Kao AH, Lacomis D, Lucas M, et al.: Anti-signal recognition particle autoantibody in patients with • and patients without idiopathic inflammatory myopathy. Arthritis Rheum 2004, 50:209– 215. [49]. Raijmakers R, Renz M, Wiemann C, et al.: PM-Scl75 is the main autoantigen in patients with the polymyositis/scleroderma overlap syndrome. Arthritis Rheum 2004, 50:565–569. SelvaOCallaghan A, MijaresBoeckhBehrens T, LabradorHorrillos M, et al.: Anti-PM-Scl antibodies in a patient with inclusion body myositis. Rheumatology 2003, 42:1016–1018. An update on pathogenesis Review (pp 700–706) Andersson J, Englund P, Sunnemark D, et al.: •• CBA/J mice infected with Trypanosoma cruzi An experimental model for inflammatory myopathies. Muscle Nerve 2003, 27:442–448. [1]. Andres N, Lizcano JM, Rodriguez MJ, et al.: Tissue activity and cellular localization of human semi• carbazide-sensitive amine oxidase. J Histochem Cytochem 2001, 49:209–217. [27]. Askanas V, Engel WK, McFerrin J, et al.: Transthyretin Val122Ile, accumulated A beta, and inclusion-body myositis aspects in cultured muscle. Neurology 2003, 61:257–260. Bassett DI, Currie PD: The zebrafish as a model for muscular dystrophy and congenital myopathy. Hum Mol Genet 2003, 12:R265–R270. Chakrabarti S, Kobayashi KS, Flavell RA, et al.: Impaired membrane resealing and autoimmune myositis in synaptotagmin VII-deficient mice. J Cell Biol 2003, 162:543–549. DeBleecker J, Vervaet V, VandenBergh P: Necrotizing myopathy with microvascular deposition of • the complement membrane attack complex. Clin Neuropathol 2004, 23:76–79. [12]. Englund P, Nennesmo I, Klareskon L, et al.: Interleukin-1alpha expression in capillaries and major histocompatibility complex class I expression in type II muscle fibers from polymyositis and dermatomyositis patients. Arthritis Rheum 2002, 46:1044–1055. Fathi M, Dastmalchi M, Rasmussen E, et al.: Interstitial lung disease, a common manifestation of • newly diagnosed polymyositis and dermatomyositis. Ann Rheum Dis 2004, 63:297–301. [7]. Fluck M, Gustafsson T: VEGF protein in human ischemic skeletal muscle. Am J Pathol 2003, 163:2636–2637. Greenberg : Molecular profiles of inflammatory myopathies (Vol 59, pg 1170, 2002). Neurology 2004, 62:678. Hino H, Araki K, Uyama E, et al.: Myopathy phenotype in transgenic mice expressing mutated PABPN1 as a model of oculopharyngeal muscular dystrophy. Hum Mol Genet 2004, 13: 181–190. Hofbauer M, Wiesener S, Babbe H, et al.: Clonal •• tracking of autoaggressive T cells in polymyositis by combining laser microdissection, single-cell PCR, and CDR3-spectratype analysis. Proc Natl Acad Sci USA 2003, 100:4090–4095. [23]. Huizing M, Rakocevic G, Sparks SE, et al.: Hypoglycosylation of alpha-dystroglycan in patients • with hereditary IBM due to GNE mutations. Molecular Genetics and Metabolism 2004, 81:196–202. [44]. Massa M, Costouros N, Mazzoli F, et al.: Self epitopes shared between human skeletal myosin and Streptococcus pyogenes M5 protein are targets of immune responses in active juvenile dermatomyositis. Arthritis Rheum 2002, 46: 3015–3025. Nakano S, Shinde A, Ito H, et al.: MAP kinase phosphatase-1 is induced in abnormal fibers in inclusion body myositis. Neurology 2003, 61: 322–326. Okada S, Weatherhead E, Targoff IN, et al.: Global •• surface ultraviolet radiation intensity may modulate the clinical and immunologic expression of autoimmune muscle disease. Arthritis Rheum 2003, 48:2285–2293. [13]. Oparil S, Zaman A, Calhoun DA: Pathogenesis of hypertension. Ann Intern Med 2003, 139:761–776. Page G, Chevrel G, Miossec P: Anatomic localization of immature and mature dendritic cell subsets in dermatomyositis and polymyositis Interaction with chemokines and Th1 cytokineproducing cells. Arthritis Rheum 2004, 50: 199–208. Pickering WP, Baker FE, Brown J, et al.: Glucocorticoid antagonist RU38486 fails to block acidinduced muscle wasting in vivo or in vitro. Nephrol Dial Transplant 2003, 18:1475–1484. Porter JD, Merriam AP, Leahy P, et al.: Temporal gene expression profiling of dystrophin-deficient (Mdx) mouse diaphragm identifies conserved and muscle group-specific mechanisms in the pathogenesis of muscular dystrophy. Hum Mol Genet 2004, 13:257– 269. Reed AM, McNallan K, Wettstein P, et al.: Does HLA-dependent chimerism underlie the pathogenesis of juvenile dermatomyositis? J Immunol 2004, 172:5041–5046. Rissanen T, YlaaHerttuala S: VEGF protein in human ischemic skeletal muscle - Author’s Reply. Am J Pathol 2003, 163:2637–2638. Schnabel A, Reuter M, Biederer J, et al.: Interstitial lung disease in polymyositis and dermatomyo• sitis - Clinical course and response to treatment. Semin Arthritis Rheum 2003, 32:273– 284. [8]. Seeliger S, Vogl T, Engels IH, et al.: Expression of calcium-binding proteins MRP8 and MRP14 in inflammatory muscle diseases. Am J Pathol 2003, 163:947–956. Seeliger S, Vogl T, Engels IH, et al.: Expression of calcium-binding proteins MRP8 and MRP14 in inflammatory muscle diseases (Vol 163, pg 947, 2003). Am J Pathol 2003, 163:2645. Tezak Z, Hoffman EP, Lutz JL, et al.: Gene expression profiling in DQA1*0501+ children with untreated dermatomyositis - A novel model of pathogenesis. J Immunol 2002, 168:4154– 4163. Torrente Y, Camirand G, Pisati F, et al.: Identification of a putative pathway for the muscle homing of stem cells in a muscular dystrophy model. J Cell Biol 2003, 162:511–520. Ulfgren AK, Grundtman C, Borg K, et al.: Downregulation of the aberrant expression of the inflammation mediator high mobility group box chromosomal protein 1 in muscle tissue of patients with polymyositis and dermatomyositis treated with corticosteroids. Arthritis Rheum 2004, 50:1586–1594. Vattemi G, Engel WK, McFerrin J, et al.: Endoplasmic reticulum stress and unfolded protein response in inclusion body myositis muscle. Am J Pathol 2004, 164:1–7. Werth VP, Callen JP, Ang G, et al.: Association of tumor necrosis factor alpha and HLA polymorphisms with adult dermatomyositis - Implications for a unique pathogenesis. J Invest Dermatol 2002, 119:617–620. Myositis and myopathies: The immunogenetics of myositis 757 Werth VP, Callen JP, Ang G, et al.: Associations of tumor necrosis factor alpha and HLA polymorphisms with adult dermatomyositis - Implications for a unique pathogenesis. J Invest Dermatol 2002, 119:617–620. Wiendl H, Mitsdoerffer M, Weller M: Express and protect yourself: The potential role of HLA-G on muscle cells and in inflammatory myopathies. Hum Immunol 2003, 64:1050–1056. Wiendl H, Mitsdoerffer M, Schneider D, et al.: Human muscle cells express a B7-related mol• ecule, B7-H1, with strong negative immune regulatory potential - A novel mechanism of counterbalancing the immune attack in idiopathic inflammatory myopathies. FASEB J 2003, 17:1892–1894. [18]. Wiendl H, Mitsdoerffer M, Schneider D, et al.: Muscle fibres and cultured muscle cells ex• press the B7.1/2-related inducible co-stimulatory molecule, ICOSL - Implications for the pathogenesis of inflammatory myopathies. Brain 2003, 126:1026–1035. [19]. Winokur ST, Chen YW, Masny PS, et al.: Expression profiling of FSHD muscle supports a defect in specific stages of myogenic differentiation. Hum Mol Genet 2003, 12:2895–2907. Yabe I, Higashi T, Kikuchi S, et al.: GNE mutations •• causing distal myopathy with rimmed vacuoles with inflammation. Neurology 2003, 61:384– 386. [45]. The immunogenetics of myositis Review (pp 707–713) Askanas V, Engel WK, McFerrin J, et al.: Transthyretin Val122Ile, accumulated Abeta, and inclu• sion-body myositis aspects in cultured muscle. Neurology 2003, 61:257–260. [41]. Bachinski LL, Udd B, Meola G, et al.: Confirmation of the type 2 myotonic dystrophy (CCTG)(N) expansion mutation in patients with proximal myotonic myopathy/proximal myotonic dystrophy of different European origins: A single shared haplotype indicates an ancestral founder effect. Am J Hum Genet 2003, 73:835– 848. Bassett DI, Currie PD: The zebrafish as a model for muscular dystrophy and congenital myopathy. Hum Mol Genet 2003, 12:R265–R270. Cagliani R, Bresolin N, Prelle A, et al.: A CAV3 microdeletion differentially affects skeletal muscle and myocardium. Neurology 2003, 61:1513–1519. FigarellaBranger D, Pouget J, Bernard R, et al.: Limb-girdle muscular dystrophy in a 71-yearold woman with an R27Q mutation in the CAV3 gene. Neurology 2003, 61:562–564. Gommans IMP, Davis M, Saar K, et al.: A locus on chromosome 15q for a dominantly inherited nemaline myopathy with core-like lesions. Brain 2003, 126:1545–1551. Gommans IMP, Davis M, Saar K, et al.: A locus on chromosome 15q for a dominantly inherited nemaline myopathy with core-like lesions (Vol 126, pg 1545, 2003). Brain 2003, 126:U11. Hassan AB, Nikitina-Zake L, Sanjeevi CB, et al.: Association of the proinflammatory haplotype • (MICA5.1/TNF2/TNFa2/DRB103) with polymyositis and dermatomyositis. Arthritis Rheum 2004, 50:1013–1015. [22]. Hino H, Araki K, Uyama E, et al.: Myopathy phenotype in transgenic mice expressing mutated PABPN1 as a model of oculopharyngeal muscular dystrophy. Hum Mol Genet 2004, 13: 181–190. Hoffman EP, Brown KJ, Eccleston E: New molecular research technologies in the study of • muscle disease. Curr Opin Rheumatol 2003, 15:698–707. [50]. Kassubek J, Juengling FD, Hoffmann S, et al.: Quantification of brain atrophy in patients with myotonic dystrophy and proximal myotonic myopathy: a controlled 3-dimensional mag- netic resonance imaging study. Neurosci Lett 2003, 348:73–76. Liquori CL, Ikeda Y, Weatherspoon M, et al.: Myotonic dystrophy type 2: Human founder haplotype and evolutionary conservation of the repeat tract. Am J Hum Genet 2003, 73:849– 862. Mahjneh I, Haravuori H, Paetau A, et al.: A distinct phenotype of distal myopathy in a large Finnish family. Neurology 2003, 61:87–92. Okada S, Weatherhead E, Targoff IN, et al.: Global •• surface ultraviolet radiation intensity may modulate the clinical and immunologic expression of autoimmune muscle disease. Arthritis Rheum 2003, 48:2285–2293. [16]. Pachman LM: Juvenile dermatomyositis - Immunogenetics, pathophysiology, and disease expression. Rheum Dis Clin North Am 2002, 28:579–592. Pachman LM: Juvenile dermatomyositis: immunogenetics, pathophysiology, and disease expression. Rheum Dis Clin North Am 2002, 28:579–602. Porter JD, Merriam AP, Leahy P, et al.: Temporal gene expression profiling of dystrophin-deficient (Mdx) mouse diaphragm identifies conserved and muscle group-specific mechanisms in the pathogenesis of muscular dystrophy. Hum Mol Genet 2004, 13:257– 269. Praud C, Montarras D, Pinset C, et al.: Dose effect relationship between the number of normal progenitor muscle cells grafted in mdx mouse skeletal striated muscle and the number of dystrophin-positive fibres. Neurosci Lett 2003, 352:70–72. Reed AM, McNallan K, Wettstein P, et al.: Does •• HLA-dependent chimerism underlie the pathogenesis of juvenile dermatomyositis? J Immunol 2004, 172:5041–5046. [33]. Stevens AM, Sullivan KM, Nelson JL: Polymyositis •• as a manifestation of chronic graft-versus-host disease. Rheumatology 2003, 42:34–39. [36]. Takahashi T, Aoki M, Tateyama M, et al.: Dysferlin mutations in Japanese Miyoshi myopathy - Relationship to phenotype. Neurology 2003, 60: 1799–1804. Tezak Z, Hoffman EP, Lutz JL, et al.: Gene expression profiling in DQA1*0501+ children with untreated dermatomyositis - A novel model of pathogenesis. J Immunol 2002, 168:4154– 4163. Tezak Z, Hoffman EP, Lutz JL, et al.: Gene expression profiling in DQA1*0501+children with untreated dermatomyositis: a novel model of pathogenesis. J Immunol 2002, 168:4154– 4163. Vihola A, Bassez G, Meola G, et al.: Histopathological differences of myotonic dystrophy type 1 (DM1) and PROMM/DM2. Neurology 2003, 60:1854–1857. Werth VP, Callen JP, Ang G, et al.: Association of tumor necrosis factor alpha and HLA polymorphisms with adult dermatomyositis - Implications for a unique pathogenesis. J Invest Dermatol 2002, 119:617–620. Werth VP, Callen JP, Ang G, et al.: Associations of tumor necrosis factor alpha and HLA polymorphisms with adult dermatomyositis - Implications for a unique pathogenesis. J Invest Dermatol 2002, 119:617–620. Wiendl H, Mitsdoerffer M, Hofmeister V, et al.: The non-classical MHC molecular HLA-G protects • human muscle cells from immune-mediated lysis. Implications for myoblast transplantation and gene therapy. Brain 2003, 126:176–185. [25]. Wiendl H, Mitsdoerffer M, Hofmeister V, et al.: The non-classical MHC molecule HLA-G protects human muscle cells from immune-mediated lysis - Implications for myoblast transplantation and gene therapy. Brain 2003, 126:176–185. Yabe I, Higashi T, Kikuchi S, et al.: GNE mutations causing distal myopathy with rimmed vacuoles with inflammation. Neurology 2003, 61:384– 386. New ideas about treatment Avramidis A, Strike PW, Taylor PN, et al.: Effectiveness of electric stimulation of the vastus medialis muscle in the rehabilitation of patients after total knee arthroplasty. Arch Phys Med Rehabil 2003, 84:1850–1853. Cherin P, Pelletier S, Teixeira A, et al.: Results and long-term follow up of intravenous immunoglobulin infusions in chronic, refractory polymyositis - An open study with thirty-five adult patients. Arthritis Rheum 2002, 46:467–474. Dalakas MC: Therapeutic approaches in patients with inflammatory myopathies. Semin Neurol 2003, 23:199–206. Lindberg C, Trysberg E, Tarkowski A, et al.: Anti-Tlymphocyte globulin treatment in inclusion body myositis - A randomized pilot study. Neurology 2003, 61:260–262. Mok CC, To CH, Szeto ML: Successful treatment of dermatomyositis-related rapidly progressive interstitial pneumonitis with sequential oral cyclophosphamide and azathioprine. Scand J Rheumatol 2003, 32:181–183. Okada S, Weatherhead E, Targoff IN, et al.: Global surface ultraviolet radiation intensity may modulate the clinical and immunologic expression of autoimmune muscle disease. Arthritis Rheum 2003, 48:2285–2293. Qu-Petersen Z, Deasy B, Jankowski R, et al.: Identification of a novel population of muscle stem cells in mice - Potential for muscle regeneration. J Cell Biol 2002, 157:851–864. Russo CR, Lauretani F, Bandinelli S, et al.: Highfrequency vibration training increases muscle power in postmenopausal women. Arch Phys Med Rehabil 2003, 84:1854–1857. Sakakima H, Yoshida Y: Effects of short duration static stretching on and reinnervated soleus muscle morphology the denervated in the rat. Arch Phys Med Rehabil 2003, 84:1339–1342. Sakkas GK, Sargeant AJ, Mercer TH, et al.: Changes in muscle morphology in dialysis patients after 6 months of aerobic exercise training. Nephrol Dial Transplant 2003, 18:1854– 1861. Song R, Lee EO, Lam P, et al.: Effects of tai chi exercise on pain, balance, muscle strength, and perceived difficulties in physical functioning in older women with osteoarthritis: A randomized clinical trial. J Rheumatol 2003, 30: 2039–2044. Sprott H, Glatzel M, Michel BA: Treatment of myositis with etanercept (Enbrel(R)), a recombinant human soluble fusion protein of TNF-alpha type II receptor and IgG1. Rheumatology 2004, 43:524–526. Stephenson J: Beefing up muscle. JAMA 2003, 290:740. Miscellaneous Abboud JA, Patel RV, DonthineniRao R, et al.: Proximal tibial segmental prosthetic replacement without the use of muscle flaps. Clin Orthop 2003, 414:189–196. Abdelgany A, Wood M, Beeson D: Allele-specific silencing of a pathogenic mutant acetylcholine receptor subunit by RNA interference. Hum Mol Genet 2003, 12:2637–2644. Agrawal S, Agrawal A, Doughty B, et al.: Cutting edge: Different toll-like receptor agonists instruct dendritic cells to induce distinct th responses via differential modulation of extracellular signal-regulated kinase-mitogen-activated protein kinase and c-fos. J Immunol 2003, 171:4984–4989. Akaogi J, Nacionales DC, Kuroda Y, et al.: Ecotropic murine leukemia viruses and exogenous 758 Myositis and myopathies: Miscellaneous mouse mammary tumor viruses are not essential for pristane-induced lupus. Arthritis Rheum 2003, 48:2990–2992. Albesiano E, Messmer BT, Damle RN, et al.: Activation-induced cytidine deaminase in chronic lymphocytic leukemia B cells: expression as multiple forms in a dynamic, variably sized fraction of the clone. Blood 2003, 102:3333– 3339. Albrand G, Munoz F, SornayRendu E, et al.: Independent predictors of all osteoporosis-related fractures in healthy postmenopausal women: The OFELY Study. Bone 2003, 32:78–85. Allanore Y, Borderie D, Lemarechal H, et al.: Acute and sustained effects of dihydropyridine-type calcium channel antagonists on oxidative stress in systemic sclerosis. Am J Med 2004, 116:595–600. Amato AA, Griggs RC: Unicorns, dragons, polymyositis, and other mythological beasts. Neurology 2003, 61:288–290. Amberg GC, Bonev AD, Rossow CF, et al.: Modulation of the molecular composition of large conductance, Ca(2+)activated K+ channels in vascular smooth muscle during hypertension. J Clin Invest 2003, 112:717–724. Anaissie EJ, Stratton SL, Dignani MC, et al.: Pathogenic molds (including Aspergillus species) in hospital water distribution sysstems: a 3-year prospective study and clinical implications for patients with hematologic malignancies. Blood 2003, 101:2542–2546. Ankarath S: Pain related to the psoas muscle after total hip replacement. J Bone Joint Surg (Br) 2003, 85B:1207. Aras RA, Fischer W, PerezPerez GI, et al.: Plasticity of repetitive DNA sequences within a bacterial (Type IV) secretion system component. J Exp Med 2003, 198:1349–1360. Arner P: Novel target genes for catecholamines in skeletal muscle. J Clin Endocrinol Metab 2004, 89:1998–1999. Aydin A, Tuncer S, Erer M, et al.: Tumoral calcinosis infiltrating the biceps brachii tendon - Excision and reconstruction with allograft: A case report. J Hand Surg [Br and Eur] 2004, 29B: 170–172. Baeten D, Kruithof E, VandenBosch F, et al.: Systematic safety follow up in a cohort of 107 patients with spondyloarthropathy treated with infliximab: a new perspective on the role of host defence in the pathogenesis of the disease? Ann Rheum Dis 2003, 62:829–834. Bakirtzis G, Choudhry R, Aasen T, et al.: Targeted epidermal expression of mutant Connexin 26(D66H) mimics true Vohwinkel syndrome and provides a model for the pathogenesis of dominant connexin disorders. Hum Mol Genet 2003, 12:1737–1744. Bakker I, Twisk JWR, vanMechelen W, et al.: Fatfree body mass is the most important body composition determinant of 10-yr longitudinal development of lumbar bone in adult men and women. J Clin Endocrinol Metab 2003, 88: 2607–2613. Ballantyne CM, Corsini A, Davidson MH, et al.: Risk for myopathy with statin therapy in high risk patients. Arch Intern Med 2003, 163:553– 564. Banga JP, Moore JK, Duhindan N, et al.: Modulation of antigen presentation by autoreactive B cell clones specific for GAD65 from a type I diabetic patient. Clin Exp Immunol 2004, 135: 74–84. Bannenberg GL, Aliberti J, Hong S, et al.: Exogenous pathogen and plant 15-lipoxygenase initiate endogenous lipoxin A4 biosynthesis. J Exp Med 2004, 199:515–523. Banu J, Wang L, Kalu DN: Effects of increased muscle mass on bone in male mice overexpressing IGF-I in skeletal muscles. Calcif Tissue Int 2003, 73:196–201. Bardoni A, Rossi P, Salvini R, et al.: Autoantibodies to RNA-polymerases in Italian patients with systemic sclerosis. Clin Exp Rheumatol 2003, 21:301–306. Bays H, Mandarino L, DeFronzo RA: Role of the adipocyte, free fatty acids, and ectopic fat in pathogenesis of type 2 diabetes mellitus: Peroxisomal proliferator-activated receptor agonists provide a rational therapeutic approach. J Clin Endocrinol Metab 2004, 89:463–478. Bearzatto M, Lampasona V, Belloni C, et al.: Fine mapping of diabetes-associated IA-2 specific autoantibodies. J Autoimmun 2003, 21:377– 382. Behrman AJ, Allan DA: Occupational exposure to bloodborne pathogens. Ann Intern Med 2004, 140:492. Beland K, Lapierre P, Marceau G, et al.: Anti-LC1 autoantibodies in patients with chronic hepatitis C virus infection. J Autoimmun 2004, 22: 159–166. Bell C, Jones PP, Seals DR: Oxidative stress does not modulate metabolic rate or skeletal muscle sympathetic activity with primary aging in adult humans. J Clin Endocrinol Metab 2003, 88: 4950–4954. Beloosesky Y, Grinblat J, Weiss A, et al.: Section II - Original articles - Pectoralis major rupture in elderly patients - A clinical study of 13 patients. Clin Orthop 2003, 413:164–169. Bernacchi E, Amato L, Parodi A, et al.: Sjogren’s syndrome: A retrospective review of the cutaneous features of 93 patients by the Italian Group of Immunodermatology. Clin Exp Rheumatol 2004, 22:55–62. Bhatti MT, Schmalfuss IM, Williams LS, et al.: Peripheral third cranial nerve enhancement in multiple sclerosis. AJNR Am J Neuroradiol 2003, 24:1390–1395. Bizzaro N, Wiik A: Appropriateness in anti-nuclear antibody testing: From clinical request to strategic laboratory practice. Clin Exp Rheumatol 2004, 22:349–355. Blank M, Krause I, Fridkin M, et al.: Baterial induction of autoantibodies to beta2-glycoprotein-I accounts for the infectious etiology of antiphospholipid syndrome. J Clin Invest 2002, 109:797–804. Bohlega S, AbuAmero SN, Wakil SM, et al.: Mutation of the slow myosin heavy chain rod domain underlies hyaline body myopathy. Neurology 2004, 62:1518–1521. Bolland MJ, Hood G, Bastin ST, et al.: Bilateral femoral head osteonecrosis after septic shock and multiorgan failure. J Bone Miner Res 2004, 19:517–520. Bona G, Defranco S, Chiocchetti A, et al.: Defective function of Fas in T cells from paediatric patients with autoimmune thyroid diseases. Clin Exp Immunol 2003, 133:430–437. Boot AM: Editorial: Body composition and bone mineral density in adolescents with partial growth hormone deficiency. J Clin Endocrinol Metab 2003, 88:5099–5100. Bourquin C, Schubart A, Tobollik S, et al.: Selective unresponsiveness to conformational B cell epitopes of the myelin oligodendrocyte glycoprotein in H-2(B) mice. J Immunol 2003, 171: 455–461. Bowen RE, Morrissy RT: Recurrence of a unicameral bone cyst in the proximal part of the fibula after en bloc resection. J Bone Joint Surg (Am) 2004, 86A:154–158. Bratke K, Bottcher B, Leeder K, et al.: Increase in granzyme B+ lymphocytes and soluble granzyme B in bronchoalveolar lavage of allergen challenged patients with atopic asthma. Clin Exp Immunol 2004, 136:542–548. Brokken LJS, Wiersinga WM, Prummel MF: Thyrotropin receptor autoantibodies are associated with continued thyrotropin suppression in treated euthyroid Graves’ disease patients. J Clin Endocrinol Metab 2003, 88:4135–4138. Bruce CR, Anderson MJ, Carey AL, et al.: Muscle oxidative capacity is a better predictor of insulin sensitivity than lipid status. J Clin Endocrinol Metab 2003, 88:5444–5451. Buchman AL, Rao SM: A patient with a polyglandular autoimmune syndrome involving the salivary glands, thyroid, intestine, and pancreas. Dig Dis Sci 2004, 49:590–593. Bujan S, OrdiRos J, Paredes J, et al.: Contribution of the initial features of systemic lupus erythematosus to the clinical evolution and survival of a cohort of Mediterranean patients. Ann Rheum Dis 2003, 62:859–865. Bushell AJ, Klenerman L, Taylor S, et al.: Ischaemic preconditioning of skeletal muscle 1. Protection against the structural changes induced by ischaemia/reperfusion injury. J Bone Joint Surg (Br) 2002, 84B:1184–1188. Bushell AJ, Klenerman L, Davies H, et al.: Ischaemic preconditioning of skeletal muscle 2. Investigation of the potential mechanisms involved. J Bone Joint Surg (Br) 2002, 84B:1189–1193. Buyon JP, Clancy RM: From antibody insult to fibrosis in neonatal lupus the heart of the matter. Arthritis Res Ther 2003, 5:266–270. Bykhovskaya Y, Casas K, Mengesha E, et al.: Missense mutation in pseudouridine synthase 1 (PUS1) causes mitochondrial myopathy and sideroblastic anemia (MLASA). Am J Hum Genet 2004, 74:1303–1308. Cabello A, Rivero A, Garcia MJ, et al.: HAART induces the expression of HLA-G on peripheral monocytes in HIV-1 infected individuals. Hum Immunol 2003, 64:1045–1049. Cairns LS, Phelps RG, Bowie L, et al.: The fine specificity and cytokine profile of T-helper cells responsive to the alpha 3 chain of type IV collagen in Goodpasture’s disease. J Am Soc Nephrol 2003, 14:2801–2812. Callaghan R: Mycobacterial knee infection in patients with idiopathic inflammatory myositis Reply. Ann Rheum Dis 2004, 63:611. Cambi A, deLange F, vanMaarseveen NM, et al.: Microdomains of the C-type lectin DC-SIGN are portals for virus entry into dendritic cells. J Cell Biol 2004, 164:145–155. Canto E, Vidal S, RodriguezSanchez JL: HKATPase expression in the susceptible BALB/c and the resistant DBA/2 strains of mice to autoimmune gastritis. Autoimmunity 2003, 36: 275–283. Carayanniotis G: The cryptic self in thyroid autoimmunity: The paradigm of thyroglobulin. Autoimmunity 2003, 36:423–428. Carlsson A, Eriksson L, Erwander I, et al.: Pseudomonas-induced lung damage in cystic fibrosis correlates to bactericidal-permeability increasing protein (BPI)-autoantibodies. Clin Exp Rheumatol 2003, 21:S95–S100. Carroccio A, Volta U, DiPrima L, et al.: Autoimmune enteropathy and colitis in an adult patient. Dig Dis Sci 2003, 48:1600–1606. Carroccio A, Iannitto E, DiPrima L, et al.: Screening for Celiac disease in non-Hodgkin’s lymphoma patients - A serum anti-transglutaminasebased approach. Dig Dis Sci 2003, 48:1530– 1536. Cha HS, Ahn KS, Jeon CH, et al.: Influence of hypoxia on the expression of matrix metalloproteinase-1,-3 and tissue inhibitor of metalloproteinase-1 in rheumatoid synovial fibroblasts. Clin Exp Rheumatol 2003, 21:593–598. Chander PN, Rocha R, Ranaudo J, et al.: Aldosterone plays a pivotal role in the pathogenesis of thrombotic microangiopathy in SHRSP. J Am Soc Nephrol 2003, 14:1990–1997. Chang M, Nakagawa PA, Williams SA, et al.: Immune thrombocytopenic purpura (ITP) plasma and purified ITP monoclonal autoantibodies inhibit megakaryocytopoiesis in vitro. Blood 2003, 102:887–895. Chauhan R, Handa R, Das TP, et al.: Over-expression of TATA binding protein (TBP) and p53 and autoantibodies to these antigens are features of systemic sclerosis, systemic lupus erythematosus and overlap syndromes. Clin Exp Immunol 2004, 136:574–584. Chazenbalk GD, Pichurin P, Chen CR: Thyroid stimulating autoantibodies in Graves’ disease Myositis and myopathies: Miscellaneous 759 preferentially recognize the free A subunit, not the thyrotropin holoreceptor. J Clin Invest 2002, 110:209–217. Chazenbalk GD, Latrofa F, McLachlan SM, et al.: Thyroid stimulation does not require antibodies with identical epitopes but does involve recognition of a critical conformation at the N terminus of the thyrotropin receptor A-subunit. J Clin Endocrinol Metab 2004, 89:1788–1793. Chen C, Lee WH, Yun P, et al.: Induction of autoantigen-specific Th2 and Tr1 regulatory T cells and modulation of autoimmune diabetes. J Immunol 2003, 171:733–744. Chen CR, Pichurin P, Nagayama Y, et al.: The thyrotropin receptor autoantigen in Graves disease is the culprit as well as the victim. J Clin Invest 2003, 111:1897–1904. Chiovato L, Latrofa F, Braverman LE, et al.: Disappearance of humoral thyroid autoimmunity after complete removal of thyroid antigens. Ann Intern Med 2003, 139:346–351. Chirinos JA, Tamariz LJ, Lopes G, et al.: Large vessel involvement in ANCA-associated vasculitides: report of a case and review of the literature. Clin Rheumatol 2004, 23:152–159. Christopoulos C, Savva S, Pylarinou S, et al.: Localised gastrocnemius myositis in Crohn’s disease. Clin Rheumatol 2003, 22:143–145. Cierny G, Mader JT, Penninck JJ: A clinical staging system for adult osteomyelitis. Clin Orthop 2003, 414:7–24. Citores MJ, RuaFigueroa I, RodriguezGallego C, et al.: The dinucleotide repeat polymorphism in the 3⬘UTR of the CD154 gene has a functional role on protein expression and is associated with systemic lupus erythematosus. Ann Rheum Dis 2004, 63:310–317. Clancy RM, Backer CB, Yin XM, et al.: Cytokine polymorphisms and histologic expression in autopsy studies: Contribution of TNF-alpha and TGF-beta 1 to the pathogenesis of autoimmune-associated congenital heart block. J Immunol 2003, 171:3253–3261. Clancy RM, Chan EKL, Buyon JP: Does the cellular localization of antigens in or on apoptotic blebs influence the pathogenicity or benefit of cognate antibodies? Comment on the article by Dieude et al. Arthritis Rheum 2003, 48: 2080–2082. Clements PJ, Becvar R, Drosos AA, et al.: Assessment of gastrointestinal involvement. Clin Exp Rheumatol 2003, 21:S15–S18. Colin EM, Uitterlinden AG, Meurs JBJ, et al.: Interaction between vitamin D receptor genotype and estrogen receptor alpha genotype influences vertebral fracture risk. J Clin Endocrinol Metab 2003, 88:3777–3784. Colino J, Snapper CM: Opposing signals from pathogen-associated molecular patterns and IL-10 are critical for optimal dendritic cell induction of in vivo humoral immunity to Streptococcus pneumoniae. J Immunol 2003, 171: 3508–3519. Collier MB, Jewett BA, Engh CA: Clinical assessment of tibial polyethylene thickness. J Arthroplasty 2003, 18:860–866. Combes V, Rosenkranz AR, Redard M, et al.: Pathogenic role of P-selectin in experimental cerebral malaria - Importance of the endothelial compartment. Am J Pathol 2004, 164:781– 786. Connolly SB, Sadlier D, Kieran NE, et al.: Transcriptome profiling and the pathogenesis of diabetic complications. J Am Soc Nephrol 2003, 14:S279–S283. Cools AM, Witvrouw EE, Declercq GA, et al.: Scapular muscle recruitment patterns: Trapezius muscle latency with and without impingement symptoms. Am J Sports Med 2003, 31: 542–549. Corti O, Hampe C, Koutnikova H, et al.: The p38 subunit of the aminoacyl-tRNA synthetase complex is a Parkin substrate: linking protein biosynthesis and neurodegeneration. Hum Mol Genet 2003, 12:1427–1437. Costerton W, Veeh R, Shirtliff M, et al.: The application of biofilm science to the study and control of chronic bacterial infections. J Clin Invest 2003, 112:1466–1477. Cremata JA, Sorell L, Montesino R, et al.: Hypogalactosylation of serum IgG in patients with coeliac disease. Clin Exp Immunol 2003, 133: 422–429. Criscione LG, Pisetsky DS: The pathogenesis of systemic lupus erythematosus. Bull Rheum Dis 2003, 52:1–6. Czaja AJ, Shums Z, Binder WL, et al.: Frequency and significance of antibodies to chromatin in autoimmune hepatitis. Dig Dis Sci 2003, 48: 1658–1664. DSilva KJ, Dwivedi AJ, Barnwell JM: Schwannoma of the psoas major muscle presenting with abdominal and back pain. Dig Dis Sci 2003, 48:1619–1621. Dahle C, Skogh T, Aberg AK, et al.: Methods of choice for diagnostic antinuclear antibody (ANA) screening benefit of adding antigenspecific assays to immunofluorescence microscopy. J Autoimmun 2004, 22:241–248. Dandona P, Aljada A, Chaudhuri A, et al.: The potential influence of inflammation and insulin resistance on the pathogenesis and treatment of atherosclerosis-related complications in type 2 diabetes. J Clin Endocrinol Metab 2003, 88:2422–2429. Daniel V, Sadeghi M, Naujokat C, et al.: Evidence for autoantibody-induced CD4 depletion mediated by apoptotic and non-apoptotic mechanisms in HIV-positive long-term surviving haemophilia patients. Clin Exp Immunol 2004, 135:94–104. Danko K, Ponyi A, Constantin T, et al.: Long-term survival of patients with idiopathic inflammatory myopathies according to clinical features - A longitudinal study of 162 cases. Medicine 2004, 83:35–42. Das H, Atsumi T, Fukushima Y, et al.: Diagnostic value of antiagalactosyl IgG antibodies in rheumatoid arthritis. Clin Rheumatol 2004, 23:218–222. Day CJ, Hewins P, Savage COS: New developments in the pathogenesis of ANCA-associated vasculitis. Clin Exp Rheumatol 2003, 21:S35–S48. DelPuente A, Esposito A, Savastano S, et al.: Rheumatoid arthritis and thyroid disease - Reply. Clin Exp Rheumatol 2004, 22:265–266. Denier C, Goutagny S, Labauge P, et al.: Mutations within the MGC4607 gene cause cerebral cavernous malformations. Am J Hum Genet 2004, 74:326–337. Deshpande N, Connelly DM, Culham EG, et al.: Reliability and validity of ankle proprioceptive measures. Arch Phys Med Rehabil 2003, 84: 883–889. Dieude M, Raymond Y, Senecal JL: Does the cellular localization of antigens in or on apoptotic blebs influence the pathogenicity or benefit of cognate antibodies? Comment on the article by Dieude et al - Reply. Arthritis Rheum 2003, 48:2082–2083. Dionne S, Laberge S, Deslandres C, et al.: Modulation of cytokine release from colonic explants by bacterial antigens in inflammatory bowel disease. Clin Exp Immunol 2003, 133:108– 114. Distler JHW, Wenger RH, Gassmann M, et al.: Physiologic responses to hypoxia and implications for hypoxia-inducible factors in the pathogenesis of rheumatoid arthritis. Arthritis Rheum 2004, 50:10–23. Dixit K, Ahsan H, Ali A: Polydeoxyribonucleotide C photoconjugated with lysine or arginine present unique epitopes for human anti-DNA autoantibodies. Hum Immunol 2003, 64:880– 886. Dixon GLJ, Heyderman RS, VanderLey P, et al.: High-level endothelial E-selectin (CD62E) cell adhesion molecule expression by a lipopolysaccharide-deficient strain of Neisseria menin- gitidis despite poor activation of NF-kappa B transcription factor. Clin Exp Immunol 2004, 135:85–93. Doi K, Hattori Y, Watanabe M: Trapezius branch of cervical plexus as donor nerve in double free muscle transfer - Reply. J Bone Joint Surg (Am) 2003, 85A:1618. Doyle HA, Gee RJ, Mamula MJ: A failure to repair self-proteins leads to T cell hyperproliferation and autoantibody production. J Immunol 2003, 171:2840–2847. Doyle CK, Davis BK, Cook RG, et al.: Hyperconservation of the N-formyl peptide binding site of M3: Evidence that M3 is an old eutherian molecule with conserved recognition of a pathogen-associated molecular pattern. J Immunol 2003, 171:836–844. Duncan GE, Perkins LA, Theriaque DW, et al.: Dichloroacetate therapy attenuates the blood lactate response to submaximal exercise in patients with defects in mitochondrial energy metabolism. J Clin Endocrinol Metab 2004, 89:1733–1738. Dwyer JP, Cohen MD: Musculoskeletal manifestations of chronic kidney disease. Bull Rheum Dis 2003, 52:1–6. DzierzanowskaFangrat K, Raeiszadeh M, Dzierzanowska D, et al.: IgG subclass response to Helicobacter pylori and CagA antigens in children. Clin Exp Immunol 2003, 134:442–446. Egawa N, Irie T, Tu YY, et al.: A case of autoimmune pancreatitis with initially negative autoantibodies turning positive during the clinical course. Dig Dis Sci 2003, 48:1705–1708. Elfving AM, Lindberg BA, Nystrom L, et al.: Islet autoantibodies in cord blood from patients who developed type 1 diabetes mellitus at 15-30 years of age. Autoimmunity 2003, 36: 227–231. Erkan D, Cervera R, Asherson RA: Catastrophic antiphospholipid syndrome - Where do we stand? Arthritis Rheum 2003, 48:3320–3327. Evoli A, Tonali PA, Padua L, et al.: Clinical correlates with anti-MuSK antibodies in generalized seronegative myasthenia gravis. Brain 2003, 126:2304–2311. Ezzat S: Editorial: Pituitary tumor pathogenesis The hunt for novel candidate genes continues. J Clin Endocrinol Metab 2003, 88:5116–5118. Fabre RA, Perez TM, Aguilar LD, et al.: Transfer factors as immunotherapy and supplement of chemotherapy in experimental pulmonary tuberculosis. Clin Exp Immunol 2004, 136:215– 223. Fagiolo E, TorianiTerenzi C: Mechanisms of immunological tolerance loss versus erythrocyte self-antigens and autoimmune hemolytic anemia. Autoimmunity 2003, 36:199–204. Fan GC, Singh RR: Vaccination with minigenes encoding V (H)-derived major histocompatibility complex class I-binding epitopes activates cytotoxic T cells that ablate autoantibody-producing B cells and inhibit lupus. J Exp Med 2002, 196:731–741. Fang CT, Chuang YP, Shun CT, et al.: A novel virulence gene in Klebsiella pneumoniae strains causing primary liver abscess and septic metastatic complications. J Exp Med 2004, 199: 697–705. Farre C, DomingoDomenech E, Font R, et al.: Celiac disease and lymphoma risk: A multicentric case-control study in Spain. Dig Dis Sci 2004, 49:408–412. Fathi M, Dastmalchi M, Rasmussen E, et al.: Interstitial lung disease, a common manifestation of newly diagnosed polymyositis and dermatomyositis. Ann Rheum Dis 2004, 63:297–301. Fava RA, Notidis E, Hunt J, et al.: A role for the lymphotoxin/LIGHT axis in the pathogenesis of murine collagen-induced arthritis. J Immunol 2003, 171:115–126. Feist E, Keitzer R, Gerhold K, et al.: Development of systemic lupus erythematosus in a patient with 760 Myositis and myopathies: Miscellaneous congenital heart block. Arthritis Rheum 2003, 48:2697–2698. Fernvik EC, Ketelhuth DFJ, Russo M, et al.: The autoantibody repertoire against copper- or macrophage-modified LDL differs in normolipidemics and hypercholesterolemic patients. J Clin Immunol 2004, 24:170–176. Ferrucci L, Guralnik JM: Inflammation, hormones, and body composition at a crossroad. Am J Med 2003, 115:501–502. Ferry H, Jones M, Vaux DJ, et al.: The cellular location of self-antigen determines the positive and negative selection of autoreactive B cells. J Exp Med 2003, 198:1415–1425. Feterowski C, Emmanuilidis K, Miethke T, et al.: Effects of functional Toll-like receptor-4 mutations on the immune response to human and experimental sepsis. Immunology 2003, 109: 426–431. Fields ML, Erikson J: The regulation of lupus-associated autoantibodies: immunoglobulin transgenic models. Curr Opin Immunol 2003, 15: 709–717. Finck BN, Han X, Courtois M, et al.: A critical role for PPARalpha-mediated lipotoxicity in the pathogenesis of diabetic cardiomyopathy: modulation by dietary fat content. Proc Natl Acad Sci USA 2003, 100:1226–1231. Fleming BC, Ohlen G, Renstrom PA, et al.: The effects of compressive load and knee joint torque on peak anterior cruciate ligament strains. Am J Sports Med 2003, 31:701–707. Fogarty PF, Rick ME, Zeng W, et al.: T cell receptor VB repertoire diversity in patients with immune thrombocytopenia following splenectomy. Clin Exp Immunol 2003, 133:461–466. Folwaczny M, Glas J, Torok HP, et al.: Toll-like receptor (TLR) 2 and 4 mutations in periodontal disease. Clin Exp Immunol 2004, 135:330– 335. Ford ML, Evavold BD: An MHC anchor-substituted analog of myelin oligodendrocyte glycoprotein 35-55 induces IFN-gamma and autoantibodies in the absence of experimental autoimmune encephalomyelitis and optic neuritis. Eur J Immunol 2004, 34:388–397. Fortin D, Salame JA, Desjardins A, et al.: Technical modification in the intracarotid chemotherapy and osmotic blood-brain barrier disruption procedure to prevent the relapse of carboplatininduced orbital pseudotumor. AJNR Am J Neuroradiol 2004, 25:830–834. Fournel S, Neichel S, Dali H, et al.: CD4(+) T cells from (New Zealand Black x New Zealand White)F-1 lupus mice and normal mice immunized against apoptotic nucleosomes recognize similar Th cell epitopes in the C terminus of histone H-3. J Immunol 2003, 171:636– 644. Frediani B, Falsetti P, Baldi F, et al.: Effects of 4-year treatment with once-weekly clodronate on prevention of corticosteroid-induced bone loss and fractures in patients with arthritis: evaluation with dual-energy X-ray absorptiometry and quantitative ultrasound. Bone 2003, 33:575–581. Freeston J, Gough A: Reversible myopathy and renal impairment. J R Soc Med 2004, 97:124– 125. Freire AD, Bertolo MB, dePinho AJ, et al.: Increased serum levels of interleukin-8 in polyarteritis nodosa and Behcet’s disease. Clin Rheumatol 2004, 23:203–205. Fuchs B, Knothe U, Hertel R, et al.: Femoral osteotomy and iliac graft vascularization for femoral head osteonecrosis. Clin Orthop 2003, 412: 84–93. Fuchtenbusch M, Bonifacio E, Lampasona V, et al.: Immune responses to glutamic acid decarboxylase and insulin in patients with gestational diabetes. Clin Exp Immunol 2004, 135:318– 321. Fujii K, Katoh S, Sairyo K, et al.: Union of defects in the pars interarticularis of the lumbar spine in children and adolescents - The radiological outcome after conservative treatment. J Bone Joint Surg (Br) 2004, 86B:225–231. Fujioka SI, Shimomura H, Iwasaki Y, et al.: Hepatitis B virus gene in liver tissue promotes hepatocellular carcinoma development in chronic hepatitis C patients. Dig Dis Sci 2003, 48: 1920–1924. Fukui M, Imamura R, Umemura M, et al.: Pathogenassociated molecular patterns sensitize macrophages to Fas ligand-induced apoptosis and IL-1 beta release. J Immunol 2003, 171:1868– 1874. Fuse K, Kodama M, Ito M, et al.: Polarity of helper T cell subsets represents disease nature and clinical course of experimental autoimmune myocarditis in rats. Clin Exp Immunol 2003, 134:403–408. Fux R, Morike K, Gundel UF, et al.: Ezetimibe and statin-associated myopathy. Ann Intern Med 2004, 140:671–672. Garchon HJ: Genetics of autoimmune myasthenia gravis, a model for antibody-mediated autoimmunity in man. J Autoimmun 2003, 21:105– 110. GeaBanacloche J, Johnson RT, Bagic A, et al.: West Nile Virus: Pathogenesis and therapeutic options. Ann Intern Med 2004, 140:545–553. Gebski BL, Mann CJ, Fletcher S, et al.: Morpholino antisense oligonucleotide induced dystrophin exon 23 skipping in mdx mouse muscle. Hum Mol Genet 2003, 12:1801–1811. Gentile F, Conte M, Formisano S: Thyroglobulin as an autoantigen: what can we learn about immunopathogenicity from the correlation of antigenic properties with protein structure? Immunology 2004, 112:13–25. Gerber LH, Furst G, Yarboro C, et al.: Number of active joints, not diagnosis, is the primary determinant of function and performance in early synovitis. Clin Exp Rheumatol 2003, 21:S65– S70. Ghirardello A, Doria A, Zampieri S, et al.: Antinucleosome antibodies in SLE: a two-year follow-up study of 101 patients. J Autoimmun 2004, 22:235–240. GiamarellosBourboulis EJ, Plachouras D, Tzivra A, et al.: Stimulation of innate immunity by susceptible and multidrug-resistant Pseudomonas aeruginosa: an in vitro and in vivo study. Clin Exp Immunol 2004, 135:240–246. Giles IP, Isenberg DA, Latchman DS, et al.: How do antiphospholipid antibodies bind beta(2)-glycoprotein I? Arthritis Rheum 2003, 48:2111– 2121. Gill DB, Koomey M, Cannon KG, et al.: Down-regulation of CD46 by piliated Neisseria gonorrhoeae. J Exp Med 2003, 198:1313–1322. Gill TJ, DeFrate LE, Wang C, et al.: The biomechanical effect of posterior cruciate ligament reconstruction on knee joint function - Kinematic response to simulated muscle loads. Am J Sports Med 2003, 31:530–536. Gillmore JD, Hutchinson WL, Herbert J, et al.: Autoimmunity and glomerulonephritis in mice with targeted deletion of the serum amyloid P component gene: SAP deficiency or strain combination? Immunology 2004, 112:255–264. Giordano R, Pellegrino M, Oleandri S, et al.: Adrenal sensitivity to adrenocorticotropin 1-24 is reduced in patients with autoimmune polyglandular syndrome. J Clin Endocrinol Metab 2004, 89:675–680. Goldfarb LG, Vicart P, Goebel HH, et al.: Desmin myopathy. Brain 2004, 127:723–734. Gora M, Gardas A, Wiktorowicz W, et al.: Evaluation of conformational epitopes on thyroid peroxidase by antipeptide antibody binding and mutagenesis. Clin Exp Immunol 2004, 136: 137–144. Goronzy JJ, Weyand CM: T-cell senescence and contraction of T-cell repertoire diversity catalysts of autoimmunity and chronic inflammation. Arthritis Res Ther 2003, 5:225–234. Gottenberg JE, Busson M, Loiseau P, et al.: In primary Sjogren’s syndrome, HLA class II is asso- ciated exclusively with autoantibody production and spreading of the autoimmune response. Arthritis Rheum 2003, 48:2240– 2245. Grandaliano G, Pontrelli P, Cerullo G, et al.: Protease-activated receptor-2 expression in IgA nephropathy: A potential role in the pathogenesis of interstitial fibrosis. J Am Soc Nephrol 2003, 14:2072–2083. Gregorio GV, Choudhuri K, Ma Y, et al.: Mimicry between the hepatitis C virus polyprotein and antigenic targets of nuclear and smooth muscle antibodies in chronic hepatitis C virus infection. Clin Exp Immunol 2003, 133:404– 413. Gris JC, Perneger TV, Quere I, et al.: Antiphospholipid/antiprotein antibodies, hemostasis-related autoantibodies, and plasma homocysteine as risk factors for a first early pregnancy loss: a matched case-control study. Blood 2003, 102:3504–3513. Groman JD, Hefferon TW, Casals T, et al.: Variation in a repeat sequence determines whether a common variant of the cystic fibrosis transmembrane conductance regulator gene is pathogenic or benign. Am J Hum Genet 2004, 74:176–179. Guarner J, Jernigan JA, Shieh WJ, et al.: Pathology and pathogenesis of bioterrorism-related inhalational anthrax. Am J Pathol 2003, 163:701– 709. Gudbjornsdottir S, Sjostrand M, Strindberg L, et al.: Direct measurements of the permeability surface area for insulin and glucose in human skeletal muscle. J Clin Endocrinol Metab 2003, 88:4559–4564. Gustavsson A, Thorsen K, Nordstrom P: A 3-year longitudinal study of the effect of physical activity on the accrual of bone mineral density in healthy adolescent males. Calcif Tissue Int 2003, 73:108–114. Gylling M, Kaariainen E, Vaisanen R, et al.: The hypoparathyroidism of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy protective effect of male sex. J Clin Endocrinol Metab 2003, 88:4602–4608. Hakkinen A, Hannonen P, Nyman K, et al.: Effects of concurrent strength and endurance training in women with early or longstanding rheumatoid arthritis: Comparison with healthy subjects. Arthritis Rheum Arthritis Care Res 2003, 49:789–797. Haller D, Holt L, Parlesak A, et al.: Differential effect of immune cells on non-pathogenic Gramnegative bacteria-induced nuclear factorkappa B activation and pro-inflammatory gene expression in intestinal epithelial cells. Immunology 2004, 112:310–320. Hammond A, Riley LH, Gailloud P, et al.: Treatment considerations for vertebroplasty in men. AJNR Am J Neuroradiol 2004, 25:639–641. Hansson AS, Johannesson M, Svensson L, et al.: Relapsing polychondritis, induced in mice with matrilin 1, is an antibody- and complementdependent disease. Am J Pathol 2004, 164: 959–966. Haq I, Isenberg D: Mycobacterial knee infection in patients with idiopathic inflammatory myositis. Ann Rheum Dis 2004, 63:611. Harmey D, Hessle L, Narisawa S, et al.: Concerted regulation of inorganic pyrophosphate and osteopontin by Akp2, Enpp1, and Ank - An integrated model of the pathogenesis of mineralization disorders. Am J Pathol 2004, 164: 1199–1209. Hedman MH, Rolandsson O, Hagg E, et al.: Association between insulin resistance and GAD65-autoantibody levels - A pilot study in an adult non-diabetic population. Autoimmunity 2004, 37:33–36. Hellmich B, Ehlers S, Csernok E, et al.: Update on the pathogenesis of Churg-Strauss syndrome. Clin Exp Rheumatol 2003, 21:S69–S77. Myositis and myopathies: Miscellaneous 761 Henneke P, Golenbock DT: Phagocytosis, innate immunity, and host-pathogen specificity. J Exp Med 2004, 199:1–4. Hicks JE, Drinkard B, Summers RM, et al.: Decreased aerobic capacity in children with juvenile dermatomyositis. Arthritis Rheum Arthritis Care Res 2002, 47:118–123. Hill JA, Southwood S, Sette A, et al.: Cutting edge: The conversion of arginine to citrulline allows for a high-affinity peptide interaction with the rheumatoid arthritis-associated HLADRB1*0401 MHC class II molecule. J Immunol 2003, 171:538–541. Hirano N, Butler MO, vonBergweltBaildon MS, et al.: Autoantibodies frequently detected in patients with aplastic anemia. Blood 2003, 102: 4567–4575. Hirata A, Masuda S, Tamura T, et al.: Expression profiling of cytokines and related genes in regenerating skeletal muscle after cardiotoxin injection - A role for osteopontin. Am J Pathol 2003, 163:203–215. Hocevar A, Tomsic M, Praprotnik S, et al.: Parasympathetic nervous system dysfunction in primary Sjogren’s syndrome. Ann Rheum Dis 2003, 62:702–704. Hoflich C, Sabat R, Rosseau S, et al.: Naturally occurring anti-IFN-gamma autoantibody and severe infections with Mycobacterium cheloneae and Burkholderia cocovenenans. Blood 2004, 103:673–675. Hoppu S, Ronkainen MS, Kulmala P, et al.: GAD65 antibody isotypes and epitope recognition during the prediabetic process in siblings of children with type I diabetes. Clin Exp Immunol 2004, 136:120–128. Horner AA, Raz E: Do microbes influence the pathogenesis of allergic diseases? Building the case for Toll-like receptor ligands. Curr Opin Immunol 2003, 15:614–619. Hsu AL, Tang PF, Jan MH: Analysis of impairments influencing gait velocity and asymmetry of hemiplegic patients after mild to moderate stroke. Arch Phys Med Rehabil 2003, 84:1185–1193. Hua J, Kumar VP, Tay SSW, et al.: Microscopic changes at the neuromuscular junction in free muscle transfer. Clin Orthop 2003, 411:325– 333. Hueber W, Utz PJ, Robinson WH: Autoantibodies in early arthritis: Advances in diagnosis and prognostication. Clin Exp Rheumatol 2003, 21:S59–S64. Humes HD: Cell therapy: Leveraging nature’s therapeutic potential. J Am Soc Nephrol 2003, 14: 2211–2213. Hummel M, Bonifacio E, Schmid S, et al.: Brief communication: Early appearance of islet autoantibodies predicts childhood type 1 diabetes in offspring of diabetic parents. Ann Intern Med 2004, 140:882–886. Hung TH, CharnockJones DS, Skepper JN, et al.: Secretion of tumor necrosis factor-alpha from human placental tissues induced by hypoxiareoxygenation causes endothelial cell activation in vitro - A potential mediator of the inflammatory response in preeclampsia. Am J Pathol 2004, 164:1049–1061. Hurley MV: Muscle dysfunction and effective rehabilitation of knee osteoarthritis: What we know and what we need to find out. Arthritis Rheum Arthritis Care Res 2003, 49:444–452. Huynh DP, Yang HT, Vakharia H, et al.: Expansion of the polyQ repeat in ataxin-2 alters its Golgi localization, disrupts the Golgi complex and causes cell death. Hum Mol Genet 2003, 12: 1485–1496. Ivashkiv LB, Hu XY: The JAK/STAT pathway in rheumatoid arthritis: Pathogenic or protective? Arthritis Rheum 2003, 48:2092–2096. Ivashkiv LB: Type I interferon modulation of cellular responses to cytokines and infectious pathogens: Potential role in SLE pathogenesis. Autoimmunity 2003, 36:473–479. Iwata A, Maruyama M, Akagi T, et al.: Alphasynuclein degradation by serine protease neurosin: implication for pathogenesis of synucleinopathies. Hum Mol Genet 2003, 12:2625– 2635. Jackson WS, TallaksenGreene SJ, Albin RL, et al.: Nucleocytoplasmic transport signals affect the age at onset of abnormalities in knock-in mice expressing polyglutamine within an ectopic protein context. Hum Mol Genet 2003, 12: 1621–1629. Jacob CO, Zang S, Li L, et al.: Pivotal role of Stat4 and Stat6 in the pathogenesis of the lupus-like disease in the New Zealand mixed 2328 mice. J Immunol 2003, 171:1564–1571. Jakkula E, Lohiniva J, Capone A, et al.: A recurrent R718W mutation in COMP results in multiple epiphyseal dysplasia with mild myopathy: clinical and pathogenetic overlap with collagen IX mutations. J Med Genet 2003, 40:942–948. Jambou F, Zhang W, Menestrier M, et al.: Circulating regulatory anti-T cell receptor antibodies in patients with myasthenia gravis. J Clin Invest 2003, 112:265–274. Jarvinen TL, Kannus P, Sievanen H: Estrogen and bone - a reproductive and locomotive perspective. J Bone Miner Res 2003, 18:1921–1931. Jarvinen TLN, Kannus P, Pajamaki I, et al.: Estrogen deposits extra mineral into bones of female rats in puberty, but simultaneously seems to suppress the responsiveness of female skeleton to mechanical loading. Bone 2003, 32: 642–651. Jasani V, WynnJones C, Richards P: Pain related to the psoas muscle after total hip replacement Reply. J Bone Joint Surg (Br) 2003, 85B:1207. Jeannet PY, Bassez G, Eymard B, et al.: Clinical and histologic findings in autosomal centronuclear myopathy. Neurology 2004, 62:1484– 1490. Jeoung D, Lim Y, Lee EB, et al.: Identification of autoantibody against poly (ADP-ribose) polymerase (PARP) fragment as a serological marker in systemic lupus erythematosus. J Autoimmun 2004, 22:87–94. Jimenez SA, Derk CT: Following the molecular pathways toward an understanding of the pathogenesis of systemic sclerosis. Ann Intern Med 2004, 140:37+. Johnson K, Svensson CI, VanEtten D, et al.: Mediation of spontaneous knee osteoarthritis by progressive chondrocyte ATP depletion in Hartley guinea pigs. Arthritis Rheum 2004, 50:1216– 1225. Jones DA, Takemoto D: Plant innate immunity - direct and indirect recognition of general and specific pathogen-associated molecules. Curr Opin Immunol 2004, 16:48–62. Jordan T, Li JY, Jiang HB, et al.: Repression of slow myosin heavy chain 2 gene expression in fast skeletal muscle fibers by muscarinic acetylcholine receptor and G alpha q signaling. J Cell Biol 2003, 162:843–850. Jorgensen TN, Gubbels MR, Kotzin BL: Links between type I interferons and the genetic basis of disease in mouse lupus. Autoimmunity 2003, 36:491–502. Jost B, Puskas GJ, Lustenberger A, et al.: Outcome of pectoralis major transfer for the treatment of irreparable subscapularis tears. J Bone Joint Surg (Am) 2003, 85A:1944–1951. Joutel A, Monet M, Domenga V, et al.: Pathogenic mutations associated with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy differently affect Jagged1 binding and Notch3 activity via the RBP/JK signaling pathway. Am J Hum Genet 2004, 74:338–347. Kamei H: Intracellular localization of histidyl-tRNA synthetase/Jo-1 antigen in T24 cells and some other cells. J Autoimmun 2004, 22:201–210. Kaneoka H, Sasatomi Y, Miyagi K, et al.: A rare case of dermatomyositis associated with immune-complex type glomerulonephritis, idio- pathic thrombopenic purpura, pulmonary fibrosis and lung cancer. Clin Exp Rheumatol 2003, 21:801–802. Karpati G, Sinnreich M: A clever road from myopathology to genes - The myotilin story. Neurology 2004, 62:1248–1249. Kasperkovitz PV, Verbeet NL, Smeets TJ, et al.: Activation of the STAT1 pathway in rheumatoid arthritis. Ann Rheum Dis 2004, 63:233–239. Kato T, Miyakawa H, Ishibashi M: Frequency and significance of anti-glutathione S-transferase autoantibody (Anti-GST A1-1) in autoimmune hepatitis. J Autoimmun 2004, 22:211–216. Kawabe S, Abe T, Kawamura H, et al.: Generation of B220(Low)B cells and production of autoantibodies in mice with experimental amyloidosis: association of primordial T cells with this phenomenon. Clin Exp Immunol 2004, 135: 200–208. Keller P, Moller K, Krabbe KS, et al.: Circulating adiponectin levels during human endotoxaemia. Clin Exp Immunol 2003, 134:107–110. Kennedy L, Evans E, Chen CM, et al.: Dramatic tissue-specific mutation length increases are an early molecular event in Huntington disease pathogenesis. Hum Mol Genet 2003, 12:3359–3367. Kerkar N, Choudhuri K, Ma Y, et al.: Cytochrome P4502D6193-212 - A new immunodominant epitope and target of virus/self cross-reactivity in liver kidney microsomal autoantibody type 1-positive liver disease. J Immunol 2003, 170: 1481–1489. Kessel A, Rosner I, Rozenbaum M, et al.: Increased CD8+ T cell apoptosis in scleroderma is associated with low levels of NF-kappa B. J Clin Immunol 2004, 24:30–36. Kifor O, McElduff A, Leboff MS, et al.: Activating antibodies to the calcium-sensing receptor in two patients with autoimmune hypoparathyroidism. J Clin Endocrinol Metab 2004, 89: 548–556. Kim HKW, Skelton DN, Quigley EJ: Pathogenesis of metaphyseal radiolucent changes following ischemic necrosis of the capital femoral epiphysis in immature pigs. J Bone Joint Surg (Am) 2004, 86A:129–135. Kimmel PL, Barisoni L, Kopp JB: Pathogenesis and treatment of HIV-associated renal diseases: Lessons from clinical and animal studies, molecular pathologic correlations, and genetic investigations. Ann Intern Med 2003, 139: 214–226. Kirman I, Jenkins D, Fowler R, et al.: Naturally occurring antibodies to epithelial cell adhesion molecule (EpCAM). Dig Dis Sci 2003, 48: 2306–2309. Knoops L, Louahed J, Renauld JC: IL-9-induced expansion of B-1b cells restores numbers but not function of B-1 lymphocytes in xid mice. J Immunol 2004, 172:6101–6106. Kobayashi Y, Murakami H, Akbar SMF, et al.: A novel and effective approach of developing aggressive experimental autoimmune gastritis in neonatal thymectomized BALB/c mouse by polyinosinic:polycytidylic acid. Clin Exp Immunol 2004, 136:423–431. Kobayashi T, Tanaka S, Okubo M, et al.: Unique epitopes of glutamic acid decarboxylase autoantibodies in slowly progressive type 1 diabetes. J Clin Endocrinol Metab 2003, 88:4768– 4775. Kohn LD, Harii N: Thyrotropin receptor autoantibodies (TSHRAbs): Epitopes, origins and clinical significance. Autoimmunity 2003, 36:331– 337. Kong PL, Zhu TP, Madaio MP, et al.: Role of the H-2 haplotype in Fas-intact lupus-prone MRL mice: association with autoantibodies but not renal disease. Arthritis Rheum 2003, 48:2992–2995. Koo TK, Mak AF, Hung LK, et al.: Joint position dependence of weakness during maximum isometric voluntary contractions in subjects with 762 Myositis and myopathies: Miscellaneous hemiparesis. Arch Phys Med Rehabil 2003, 84:1380–1386. Kovacs L, Papos M, Takacs R, et al.: Autonomic nervous system dysfunction involving the gastrointestinal and the urinary tracts in primary Sjogren’s syndrome. Clin Exp Rheumatol 2003, 21:697–703. Koya D, Hayashi K, Kitada M, et al.: Effects of antioxidants in diabetes-induced oxidative stress in the glomeruli of diabetic rats. J Am Soc Nephrol 2003, 14:S250–S253. Kralovics R, Stockton DW, Prchal JT: Clonal hematopoiesis in familial polycythemia vera suggests the involvement of multiple mutational events in the early pathogenesis of the disease. Blood 2003, 102:3793–3796. Kraus VB, Stabler T, Le ET, et al.: Urinary type II collagen neoepitope as an outcome measure for relapsing polychondritis. Arthritis Rheum 2003, 48:2942–2948. Kumar A, Mann S, Sossi V, et al.: [C-11]DTBZ-PET correlates of levodopa responses in asymmetric Parkinson’s disease. Brain 2003, 126:2648–2655. Kunigal S, Kusch A, Tkachuk N, et al.: Monocyteexpressed urokinase inhibits vascular smooth muscle cell growth by activating Stat1. Blood 2003, 102:4377–4383. Kuroki A, Moll T, LopezHoyos M, et al.: Enforced Bcl-2 expression in B lymphocytes induces rheumatoid factor and anti-DNA production, but the Yaa mutation promotes only anti-DNA production. Eur J Immunol 2004, 34:1077– 1084. Kuroki T, Isshiki K, King GL: Oxidative stress: The lead or supporting actor in the pathogenesis of diabetic complications. J Am Soc Nephrol 2003, 14:S216–S220. Kusakabe N, Sekiya S, Harada T, et al.: Trapezius branch of cervical plexus as donor nerve in double free muscle transfer. J Bone Joint Surg (Am) 2003, 85A:1617–1618. LaMontagna G, Meli R, Criscuolo T, et al.: Bioactivity of prolactin in systemic sclerosis. Clin Exp Rheumatol 2004, 22:145–150. Lamb SE, Frost H: Recovery of mobility after knee arthroplasty - Expected rates and influencing factors. J Arthroplasty 2003, 18:575–582. Lambert CP, Archer RL, Carrithers JA, et al.: Influence of creatine monohydrate ingestion on muscle metabolites and intense exercise capacity in individuals with multiple sclerosis. Arch Phys Med Rehabil 2003, 84:1206–1210. Lang TJ, Nguyen P, Papadimitriou JC, et al.: Increased severity of murine lupus in female mice is due to enhanced expansion of pathogenic T cells. J Immunol 2003, 171:5795– 5801. Langguth DM, Wong RCW, Archibald C, et al.: Haemorrhagic myositis associated with prophylactic heparin use in dermatomyositis. Ann Rheum Dis 2004, 63:464–465. Lard LR, vanGaalen FA, Schonkeren JJM, et al.: Association of the-2849 interleukin-10 promoter polymorphism with autoantibody production and joint destruction in rheumatoid arthritis. Arthritis Rheum 2003, 48:1841–1848. Latham KA, Zamora A, Drought H, et al.: Estradiol treatment redirects the isotype of the autoantibody response and prevents the development of autoimmune arthritis. J Immunol 2003, 171: 5820–5827. Laurie KL, LaGruta NL, Koch N, et al.: Thymic expression of a gastritogenic epitope results in positive selection of self-reactive pathogenic T cells. J Immunol 2004, 172:5994–6002. Lauvau G, Glaichenhaus N: Presentation of pathogen-derived antigens in vivo. Eur J Immunol 2004, 34:913–920. Layland LE, Wulferink M, Dierkes S, et al.: Druginduced autoantibody formation in mice: triggering by primed CD4(+)CD25(−) T cells, prevention by primed CD4(+)CD25(+) T cells. Eur J Immunol 2004, 34:36–46. Lee DM, Schur PH: Clinical utility of the anti-CCP assay in patients with rheumatic diseases. Ann Rheum Dis 2003, 62:870–874. Lee KH, Chung HS, Kim HS, et al.: Human alphaenolase from endothelial cells as a target antigen of anti-endothelial cell antibody in Behcet’s disease. Arthritis Rheum 2003, 48:2025–2035. Lee SW, Lee HJ, Chung WT, et al.: TRAIL induces apoptosis of chondrocytes and influences the pathogenesis of experimentally induced rat osteoarthritis. Arthritis Rheum 2004, 50:534– 542. Leppilahti M, Tammela TLJ, Huhtala H, et al.: Interstitial cystitis-like urinary symptoms among patients with Sjogren’s syndrome: A population-based study in Finland. Am J Med 2003, 115:62–65. Lester S, Danda D, Cavill D, et al.: Humoral immunity to Ro52 is not associated with the Ro52 9571 C/T polymorphism in Australian patients with primary Sjogren’s syndrome. Arthritis Rheum 2003, 48:3293–3294. Leung DYM, Jain N, Leo HL: New concepts in the pathogenesis of atopic dermatitis. Curr Opin Immunol 2003, 15:634–638. Levidiotis V, Freeman C, Tikellis C, et al.: Heparanase is involved in the pathogenesis of proteinuria as a result of glomerulonephritis. J Am Soc Nephrol 2004, 15:68–78. Li N, Nakamura K, Jiang Y, et al.: Gain-of-function polymorphism in mouse and human Ltk: implications for the pathogenesis of systemic lupus erythematosus. Hum Mol Genet 2004, 13: 171–179. Li XD, Cui QJ, Kao CH, et al.: Lovastatin inhibits adipogenic and stimulates osteogenic differentiation by suppressing PPAR gamma 2 and increasing Cbfa1/Runx2 expression in bone marrow mesenchymal cell cultures. Bone 2003, 33:652–659. Li AH, Zhou JB, Zuckerman D, et al.: Sequence analysis of clonal immunoglobulin and T-cell receptor gene rearrangements in children with acute lymphoblastic leukemia at diagnosis and at relapse: implications for pathogenesis and for the clinical utility of PCR-based methods of minimal residual disease detection. Blood 2003, 102:4520–4526. Li N, Aoki V, Hans G, et al.: The role of intramolecular epitope spreading in the pathogenesis of endemic pemphigus foliaceus (Fogo Selvagem). J Exp Med 2003, 197:1501–1510. Li SH, VandenDiepstraten C, DSouza SJ, et al.: Vascular smooth muscle cells orchestrate the assembly of type I collagen via alpha 2 beta 1 integrin, RhoA, and fibronectin polymerization. Am J Pathol 2003, 163:1045–1056. Liang ZY, Xie C, Chen C, et al.: Pathogenic profiles and molecular signatures of antinuclear autoantibodies rescued from NZM2410 lupus mice. J Exp Med 2004, 199:381–398. Licht R, Dieker JWC, Jacobs CWM, et al.: Decreased phagocytosis of apoptotic cells in diseased SLE mice. J Autoimmun 2004, 22: 139–145. Lieby P, Soley A, Knapp AM, et al.: Memory B cells producing somatically mutated antiphospholipid antibodies are present in healthy individuals. Blood 2003, 102:2459–2465. Lily O, Palace J, Vincent A: Serum autoantibodies to cell surface determinants in multiple sclerosis: a flow cytometric study. Brain 2004, 127: 269–279. Lim SY, Ghosh SK: Autoreactive responses to an environmental factor. 2. Phthalate-induced anti-DNA specificity is downregulated by autoreactive cytotoxic T cells. Immunology 2004, 112:94–104. Lim SY, Ghosh SK: Autoreactive responses to an environmental factor: 1. phthalate induces antibodies exhibiting anti-DNA specificity. Immunology 2003, 110:482–492. Lim VS, Yarasheski KE, Crowley JR, et al.: Insulin is protein-anabolic in chronic renal failure patients. J Am Soc Nephrol 2003, 14:2297– 2304. Liquori CL, Berg MJ, Siegel AM, et al.: Mutations in a gene encoding a novel protein containing a phosphotyrosine-binding domain cause type 2 cerebral cavernous malformations. Am J Hum Genet 2003, 73:1459–1464. Liu DX, Cai SH, Gu XG, et al.: C1 inhibitor prevents endotoxin shock via a direct interaction with lipopolysaccharide. J Immunol 2003, 171:2594–2601. Liu W, Thomas SG, Asa SL, et al.: Myostatin is a skeletal muscle target of growth hormone anabolic action. J Clin Endocrinol Metab 2003, 88:5490–5496. Liu E, Morlyama H, Paronen J, et al.: Nondepleting anti-CD4 monoclonal antibody prevents diabetes and blocks induction of insulin autoantibodies following insulin peptide B:9-23 immunization in the NOD mouse. J Autoimmun 2003, 21:213–219. Loizou S, Singh S, Wypkerna E, et al.: Anticardiolipin, anti-beta(2)-glycoprotein I and antiprothrombin antibodies in black South African patients with infectious disease. Ann Rheum Dis 2003, 62:1106–1111. Loria P, Lonardo A, Leonardi F, et al.: Non-organspecific autoantibodies in nonalcoholic fatty liver disease: Prevalence and correlates. Dig Dis Sci 2003, 48:2173–2181. Lotscher M, Recher M, Hunziker L, et al.: Immunologically induced, complement-dependent upregulation of the prion protein in the mouse spleen: Follicular dendritic cells versus capsule and trabeculae. J Immunol 2003, 170: 6040–6047. Lou YH, Borillo J: Migration of T cells from nearby inflammatory foci into antibody bound tissue: a relay of T cell and antibody actions in targeting native autoantigen. J Autoimmun 2003, 21: 27–35. Lugering A, Floer M, Lugering N, et al.: Characterization of M cell formation and associated mononuclear cells during indomethacin-induced intestinal inflammation. Clin Exp Immunol 2004, 136:232–238. Lutz J, Fedczyna T, Huwiler K, et al.: Increased plasma thrombospondin-1 (TSP-1) levels are associated with the TNFalpha-308A allele in children with juvenile dermatomyositis (JDM). Clin Immunol 2003, 103:250–263. MaKrupa W, Jeon MS, Spoerl S, et al.: Activation of arterial wall dendritic cells and breakdown of self-tolerance in giant cell arteritis. J Exp Med 2004, 199:173–183. Macdonald P, Palmer J, Kirby JA, et al.: Apoptosis as a mechanism for cell surface expression of the autoantigen pyruvate dehydrogenase complex. Clin Exp Immunol 2004, 136:559–567. Maes B, Demunter A, Peeters B, et al.: BCL10 mutation does not represent an important pathogenic mechanism in gastric MALT type lymphoma, and the presence of the API2-MLT fusion is associated with aberrant nuclear BCL10 expression. Blood 2002, 99:1398– 1404. Magrangeas F, Nasser V, AvetLoiseau H, et al.: Gene expression profiling of multiple myeloma reveals molecular portraits in relation to the pathogenesis of the disease. Blood 2003, 101:4998–5006. Mahler E, Hoebeke J, Levin MJ: Structural and functional complexity of the humoral response against the Trypanosoma cruzi ribosomal P2 beta protein in patients with chronic Chagas’ heart disease. Clin Exp Immunol 2004, 136: 527–534. Manavalan SJ, Valiando JR, Reeves WH, et al.: Genomic absence of the gene encoding T cell receptor V(Beta)7.2 is linked to the presence of autoantibodies in Sjogren’s syndrome. Arthritis Rheum 2004, 50:187–198. Myositis and myopathies: Miscellaneous 763 Mannerkorpi K, Iversen MD: Physical exercise in fibromyalgia and related syndromes. Best Pract Res Clin Rheumatol 2003, 17:629–647. Mannik M: Use of novel elution regimens of autoantibodies in lupus kidneys: comment on the article by Xie et al. Arthritis Rheum 2004, 50: 1699–1700. Manno : AAV-mediated factor IX gene transfer to skeletal muscle in patients with severe hemophilia B (Vol 101, pg 2963, 2003). Blood 2003, 102:788. Mantovani G, Maghnie M, Weber G, et al.: Growth hormone-releasing hormone resistance in pseudohypoparathyroidism type Ia: New evidence for imprinting of the Gs alpha gene. J Clin Endocrinol Metab 2003, 88:4070–4074. Marie I, Fournet P, Janvresse A, et al.: Periarticular calcifications and arthropathy as the first manifestation of polymyositis. Clin Exp Rheumatol 2003, 21:681–682. Marquina R, Diez MA, LopezHoyos M, et al.: Inhibition of B cell death causes the development of an IgA nephropathy in (New Zealand white x C57BL/6)F-1-bcl-2 transgenic mice. J Immunol 2004, 172:7177–7185. Maruyama K, Muramatsu H, Ishiguro N, et al.: Midkine, a heparin-binding growth factor, is fundamentally involved in the pathogenesis of rheumatoid arthritis. Arthritis Rheum 2004, 50:1420–1429. Matarrese P, Gambardella L, Cassone A, et al.: Mitochondrial membrane hyperpolarization hijacks activated T lymphocytes toward the apoptotic-prone phenotype: Homeostatic mechanisms of HIV protease inhibitors. J Immunol 2003, 170:6006–6015. Mathieson PW: What has the immune system got against the glomerular podocyte? Clin Exp Immunol 2003, 134:1–5. Matsumoto M, Kokame K, Soejima K, et al.: Molecular characterization of ADAMTS13 gene mutations in Japanese patients with Upshaw-Schulman syndrome. Blood 2004, 103:1305–1310. Maury CP, Liljestrom M, Laiho K, et al.: Tumor necrosis factor alpha, its soluble receptor I, and 308 gene promoter polymorphism in patients with rheumatoid arthritis with or without amyloidosis: implications for the pathogenesis of nephropathy and anemia of chronic disease in reactive amyloidosis. Arthritis Rheum 2003, 48:3068–3076. Mayne M, Moffatt T, Kong H, et al.: CYFIP2 is highly abundant in CD4(+) cells from multiple sclerosis patients and is involved in T cell adhesion. Eur J Immunol 2004, 34:1217–1227. McGarvey JA, Wagner D, Bermudez LE: Differential gene expression in mononuclear phagocytes infected with pathogenic and non-pathogenic mycobacteria. Clin Exp Immunol 2004, 136: 490–500. McHugh NJ, Distler O, Giacomelli R, et al.: Non organ based laboratory markers in systemic sclerosis. Clin Exp Rheumatol 2003, 21:S32– S38. Meffre E, Schaefer A, Wardemann H, et al.: Surrogate light chain expressing human peripheral B cells produce self-reactive antibodies. J Exp Med 2004, 199:145–150. Mehta S, Gimbel JA, Soslowsky LJ: Etiologic and pathogenetic factors for rotator cuff tendinopathy. Clin Sports Med 2003, 22:791+. Melanitou E, Fain P, Eisenbarth GS: Genetics of Type 1A (Immune mediated) diabetes. J Autoimmun 2003, 21:93–98. Mendez EP, Lipton R, RamseyGoldman R, et al.: US incidence of juvenile dermatomyositis, 1995-1998: Results from the National Institute of Arthritis and Musculoskeletal and Skin Diseases Registry. Arthritis Rheum Arthritis Care Res 2003, 49:300–305. Mennechet FJD, Kasper LH, Rachinel N, et al.: Intestinal intraepithelial lymphocytes prevent pathogen-driven inflammation and regulate the Smad/T-bet pathway of lamina propria CD4(+) T cells. Eur J Immunol 2004, 34:1059–1067. MeyerBahlburg A, Greil S, Kruse N, et al.: Yersinia enterocolitica leads to transient induction of TNF-alpha and activates NF-kappa B in synovial fibroblasts. Clin Exp Rheumatol 2004, 22: 278–284. Michalik A, VanBroeckhoven C: Pathogenesis of polyglutamine disorders: aggregation revisited. Hum Mol Genet 2003, 12:R173–R186. Millet ACM, Ewbank JJ: Immunity in Caenorhabditis elegans. Curr Opin Immunol 2004, 16:4–9. Minich WB, Lenzner C, Bergmann A, et al.: A coated tube assay for the detection of blocking thyrotropin receptor autoantibodies. J Clin Endocrinol Metab 2004, 89:352–356. Minich WB, Lenzner C, Morgenthaler NG: Antibodies to TSH-receptor in thyroid autoimmune disease interact with monoclonal antibodies whose epitopes are broadly distributed on the receptor. Clin Exp Immunol 2004, 136:129– 136. Miyachi K, Hirano Y, Horigome T, et al.: Autoantibodies from primary biliary cirrhosis patients with anti-p95c antibodies bind to recombinant p97/VCP and inhibit in vitro nuclear envelope assembly. Clin Exp Immunol 2004, 136:568– 573. Miyakawa H, Kawashima Y, Kitazawa E, et al.: Low frequency of anti-SLA/LP autoantibody in Japanese adult patients with autoimmune liver disease - Analysis with recombinant antigen assay. J Autoimmun 2003, 21:77–82. Modlin RL, Doherty P: Host-pathogen interactions Host defense against microbial pathogens the immune system’s weapons of mass destruction. Curr Opin Immunol 2003, 15:393– 395. Moghadaszadeh B, Albrechtsen R, Guo LT, et al.: Compensation for dystrophin-deficiency: ADAM12 overexpression in skeletal muscle results in increased alpha 7 integrin, utrophin and associated glycoproteins. Hum Mol Genet 2003, 12:2467–2479. Mohan C, Xie C, Liang ZY, et al.: Use of novel elution regimens of autoantibodies in lupus kidneys: comment on the article by Xie et al - Reply. Arthritis Rheum 2004, 50:1700. Mok CC, Tsui EYK, Chau SY: Erosive arthropathy in amyopathic dermatomyositis. Clin Exp Rheumatol 2003, 21:409–410. Mongini PKA, Jackson AE, Tolani S, et al.: Role of complement-binding CD21/CD19/CD81 in enhancing human B cell protection from fasmediated apoptosis. J Immunol 2003, 171: 5244–5254. Montana ES, Littleton JT: Characterization of a hypercontraction-induced myopathy in Drosophila caused by mutations in Mhc. J Cell Biol 2004, 164:1045–1054. Morabito N, Gaudio A, Lasco A, et al.: Osteoprotegerin and RANKL in the pathogenesis of thalassemia-induced osteoporosis: new pieces of the puzzle. J Bone Miner Res 2004, 19:722– 727. Moreland JD, Goldsmith CH, Huijbregts MP, et al.: Progressive resistance strengthening exercises after stroke: A single-blind randomized controlled trial. Arch Phys Med Rehabil 2003, 84:1433–1440. Moschetta A, Twickler TB, Rehfeld JF, et al.: Effects of growth hormone deficiency and recombinant growth hormone therapy on postprandial gallbladder motility and cholecystokinin release. Dig Dis Sci 2004, 49:529–534. Mrena S, Savola K, Kulmala P, et al.: Genetic modification of risk assessment based on staging of preclinical type 1 diabetes in siblings of affected children. J Clin Endocrinol Metab 2003, 88:2682–2689. Murashima A, Fukazawa T, Hirashima M, et al.: Long term prognosis of children born to lupus patients. Ann Rheum Dis 2004, 63:50–53. Muratori P, Muratori L, Gershwin ME, et al.: ’True’ antimitochondrial antibody-negative primary biliary cirrhosis, low sensitivity of the routine assays, or both? Clin Exp Immunol 2004, 135: 154–158. Muratori P, Muratori L, Guidi M, et al.: Anti-Saccharomyces cerevisiae antibodies (ASCA) and autoimmune liver diseases. Clin Exp Immunol 2003, 132:473–476. Murphy E, Freaney R, Bresnihan B, et al.: Increased bone resorption and failure to respond to antiresorptive therapy in progressive dystrophic calcification. Calcif Tissue Int 2003, 73:433– 440. Nagayama Y, Mizuguchi H, Hayakawa T, et al.: Prevention of autoantibody mediated Graves’-like hyperthyroidism in mice by interleukin-4, a Th2 cytokine. J Immunol 2003, 170:3522–3527. Naito T, Ogasawara H, Kaneko H, et al.: Immune abnormalities induced by human endogenous retroviral peptides: with reference to the pathogenesis of systemic lupus erythematosus. J Clin Immunol 2003, 23:371–376. Nakachi K, Powell M, Swift G, et al.: Epitopes recognised by tissue transglutaminase antibodies in coeliac disease. J Autoimmun 2004, 22:53–63. Nakamura A, Funatomi H, Katagiri A, et al.: A case of autoimmune pancreatitis complicated with immune thrombocytopenia during maintenance therapy with prednisolone. Dig Dis Sci 2003, 48:1968–1971. Nakamura Y, Wakitani S, Nakayama J, et al.: Temporal and spatial expression profiles of BMP receptors and Noggin during BMP-2-induced ectopic bone formation. J Bone Miner Res 2003, 18:1854–1862. Nally JE, Chantranuwat C, Wu XY, et al.: Alveolar septal deposition of immunoglobulin and complement parallels pulmonary hemorrhage in a guinea pig model of severe pulmonary leptospirosis. Am J Pathol 2004, 164:1115– 1127. Napoli R, Guardasole V, Angelini V, et al.: Acute effects of growth hormone on vascular function in human subjects. J Clin Endocrinol Metab 2003, 88:2817–2820. Nicolo D, Goldman BI, Monestier M: The antiphospholipid syndrome and atherosclerosis: clue to pathogenesis. Curr Rheumatol Rep 2003, 5: 401–406. Niederau C: Do cholecystokinin antagonists increase cytosolic calcium in pancreatic acinar cells and thereby promote pancreatitis? Dig Dis Sci 2004, 49:266–269. Nielen MMJ, vanSchaardenburg D, Reesink HW, et al.: Specific autoantibodies precede the symptoms of rheumatoid arthritis - A study of serial measurements in blood donors. Arthritis Rheum 2004, 50:380–386. Niiyama H, Jagannath S, Kantsevoy S, et al.: Intrasphincteric nitric oxide reduces sphincter of Oddi motility in an endoscopic porcine model. Dig Dis Sci 2003, 48:2187–2190. Nili N, Cheema AN, Giordano FJ, et al.: Decorin inhibition of PDGF-stimulated vascular smooth muscle cell function - Potential mechanism for inhibition of intimal hyperplasia after balloon angioplasty. Am J Pathol 2003, 163:869–878. Noble A, Leggat JA, Inderberg EM: CD8(+) immunoregulatory cells in the graft-versus-host reaction: CD8 T cells activate dendritic cells to secrete interleukin-12/interleukin-18 and induce T helper I autoantibody. Immunology 2003, 109:476–486. Nollet F, Beelen A, Twisk JW, et al.: Perceived health and physical functioning in postpoliomyelitis syndrome: A 6-year prospective follow-up study. Arch Phys Med Rehabil 2003, 84:1048–1056. Norris AW, Chen LH, Fisher SJ, et al.: Muscle-specific PPAR gamma-deficient mice develop increased adiposity and insulin resistance but respond to thiazolidinediones. J Clin Invest 2003, 112:608–618. Nosadini R, Tonolo G: Relationship between blood glucose control, pathogenesis and progres- 764 Myositis and myopathies: Miscellaneous sion of diabetic nephropathy. J Am Soc Nephrol 2004, 15:S1–S5. Oppezzo P, Dumas G, Bouvet JP, et al.: Somatic mutations can lead to a loss of superantigenic and polyreactive binding. Eur J Immunol 2004, 34:1423–1432. OrtizUrda S, ElbeBurger A, Smolle J, et al.: The plant lectin wheat germ agglutinin inhibits the binding of pemphigus foliaceus autoantibodies to desmoglein 1 in a majority of patients and prevents pathomechanisms of pemphigus foliaceus in vitro and in vivo. J Immunol 2003, 171:6244–6250. Otsuka J, Horiuchi T, Yoshizawa S, et al.: Association of a four-amino acid residue insertion polymorphism of the HS1 gene with systemic lupus erythematosus - Molecular and functional analysis. Arthritis Rheum 2004, 50:871–881. Ott PA, Dittrich MT, Herzog BA, et al.: T cells recognize multiple GAD65 and proinsulin Epitopes in human type 1 diabetes, suggesting determinant spreading. J Clin Immunol 2004, 24:327–339. Ou Y, Enarson P, Rattner JB, et al.: The nuclear pore complex protein Tpr is a common autoantigen in sera that demonstrate nuclear envelope staining by indirect immunofluorescence. Clin Exp Immunol 2004, 136:379–387. Pachlopnik JM, Horn MP, Fux M, et al.: Natural antiFc epsilon RIa autoantibodies may interfere with diagnostic tests for autoimmune urticaria. J Autoimmun 2004, 22:43–51. Palmieri RM, Ingersoll CD, Edwards JE, et al.: Arthrogenic muscle inhibition is not present in the limb contralateral to a simulated knee joint effusion. Am J Phys Med Rehabil 2003, 82:910–916. Papadonikolakis AS, Vekris MD, Korompilias AV, et al.: Botulinum A toxin for treatment of lower limb spasticity in cerebral palsy - Gait analysis in 49 patients. Acta Orthop Scand 2003, 74: 749–755. Papamattheou MG, Routsias JG, Karagouni EE, et al.: T cell help is required to induce idiotypicanti-idiotypic autoantibody network after immunization with complementary epitope 289308aa of La/SSB autoantigen in nonautoimmune mice. Clin Exp Immunol 2004, 135:416–426. Park HS, Francis KP, Yu J, et al.: Membranous cells in nasal-associated lymphoid tissue: A portal of entry for the respiratory mucosal pathogen group A streptococcus. J Immunol 2003, 171: 2532–2537. Patry YC, Nachman PH, Audrain MAP, et al.: Difference in antigenic determinant profiles between human and rat myeloperoxidase. Clin Exp Immunol 2003, 132:505–508. Pearce SHS, Leech NJ: Editorial: Toward precise forecasting of autoimmune endocrinopathy. J Clin Endocrinol Metab 2004, 89:544–547. Pellicano R, Touscoz GA, Smedile A, et al.: Prevalence of non-organ-specific autoantibodies in patients suffering from duodenal ulcer with and without Helicobacter pylori infection. Dig Dis Sci 2004, 49:395–398. Peng B, Wu W, Hou S, et al.: The pathogenesis of Schmorl’s nodes. J Bone Joint Surg (Br) 2003, 85B:879–882. Persson AL, Hansson GA, Kalliomaki J, et al.: Increases in local pressure pain thresholds after muscle exertion in women with chronic shoulder pain. Arch Phys Med Rehabil 2003, 84: 1515–1522. Pichurin PN, Chen CR, Nagayama Y, et al.: Evidence that factors other than particular thyrotropin receptor T cell epitopes contribute to the development of hyperthyroidism in murine Graves’ disease. Clin Exp Immunol 2004, 135: 391–397. Pichurin P, Aliesky H, Chen CR, et al.: Thyrotrophin receptor-specific memory T cell responses require normal B cells in a murine model of Graves’ disease. Clin Exp Immunol 2003, 134: 396–402. Pichyangkul S, Endy TP, Kalayanarooj S, et al.: A blunted blood plasmacytoid dendritic cell response to an acute systemic viral infection is associated with increased disease severity. J Immunol 2003, 171:5571–5578. Pierangeli SS, Blank M, Liu XW, et al.: A peptide that shares similarity with bacterial antigens reverses thrombogenic properties of antiphospholipid antibodies in vivo. J Autoimmun 2004, 22:217–225. Pittoni V, Sorice M, Circella A, et al.: Specificity of antinuclear and antiphospholipid antibodies in sera of patients with autoimmune lymphoproliferative disease (ALD). Clin Exp Rheumatol 2003, 21:377–385. Porter JD, Merriam AP, Leahy P, et al.: Dissection of temporal gene expression signatures of affected and spared muscle groups in dystrophin-deficient (Mdx) mice. Hum Mol Genet 2003, 12:1813–1821. Pullmann R, Skerenova M, Lukac J, et al.: No role of factor V Leiden and prothrombin G20210A mutations in the pathogenesis of atherosclerosis arterial thrombosis in SLE. Clin Exp Rheumatol 2004, 22:138–139. Qian Q, Li M, Cai YQ, et al.: Analysis of the polycystins in aortic vascular smooth muscle cells. J Am Soc Nephrol 2003, 14:2280–2287. Qian ZR, Sano T, Asa SL, et al.: Cytoplasmic expression of fibroblast growth factor receptor-4 in human pituitary adenomas: Relation to tumor type, size, proliferation, and invasiveness. J Clin Endocrinol Metab 2004, 89:1904–1911. Qian Q, Hunter LW, Li M, et al.: Pkd2 haploinsufficiency alters intracellular calcium regulation in vascular smooth muscle cells. Hum Mol Genet 2003, 12:1875–1880. Qiu SJ, Rao DS, Palnitkar S, et al.: Reduced iliac cancellous osteocyte density in patients with osteoporotic vertebral fracture. J Bone Miner Res 2003, 18:1657–1663. Quintana FJ, Getz G, Hed G, et al.: Cluster analysis of human autoantibody reactivities in health and in type 1 diabetes mellitus: a bio-informatic approach to immune complexity. J Autoimmun 2003, 21:65–75. Rader DJ, Cohen J, Hobbs HH: Monogenic hypercholesterolemia - New insights in pathogenesis and treatment. J Clin Invest 2003, 111: 1795–1803. Rader DJ, Cohen J, Hobbs HH: Monogenic hypercholesterolemia: new insights in pathogenesis and treatment. J Clin Invest 2003, 111:1795– 1803. RamirezPineda JR, Frohlich A, Berberich C, et al.: Dendritic cells (DC) activated by CpG DNA ex vivo are potent inducers of host resistance to an intracellular pathogen that is independent of IL-12 derived from the immunizing DC. J Immunol 2004, 172:6281–6289. Rand JH, Wu XX, Quinn AS, et al.: Human monoclonal antiphospholipid antibodies disrupt the annexin A5 anticoagulant crystal shield on phospholipid bilayers - Evidence from atomic force microscopy and functional assay. Am J Pathol 2003, 163:1193–1200. Ranum LPW, Day JW: Myotonic dystrophy: RNA pathogenesis comes into focus. Am J Hum Genet 2004, 74:793–804. Rao RD, Singrakhia MD: Painful osteoporotic vertebral fracture - Pathogenesis, evaluation, and roles of vertebroplasty and kyphoplasty in its management. J Bone Joint Surg (Am) 2003, 85A:2010–2022. Raupach B, Kurth N, Pfeffer K, et al.: Salmonella typhimurium strains carrying independent mutations display similar virulence phenotypes yet are controlled by distinct host defense mechanisms. J Immunol 2003, 170:6133–6140. Reiners KS, Hansen HP, Krussmann A, et al.: Selective killing of B-cell hybridomas targeting proteinase 3, Wegener’s autoantigen. Immunology 2004, 112:228–236. Remer T, Boye KR, Hartmann M, et al.: Adrenarche and bone modeling and remodeling at the proximal radius: Weak androgens make stronger cortical bone in healthy children. J Bone Miner Res 2003, 18:1539–1546. Reumaux D, Duthilleul P, Roos D: Pathogenesis of diseases associated with antineutrophil cytoplasm autoantibodies. Hum Immunol 2004, 65:1–12. Riente L, Bongiorni F, Nacci A, et al.: Antibodies to inner ear antigens in Meniere’s disease. Clin Exp Immunol 2004, 135:159–163. Roman RMB, Lobo PI, Taylor RP, et al.: Prospective study of the immune effects of normalizing the hemoglobin concentration in hemodialysis patients who receive recombinant human erythropoietin. J Am Soc Nephrol 2004, 15:1339– 1346. Romano CL, Duci D, Romano D, et al.: Celecoxib versus indomethacin in the prevention of heterotopic ossification after total hip arthroplasty. J Arthroplasty 2004, 19:14–18. Rome S, Clement K, Rabasa-Lhoret R, et al.: Microarray profiling of human skeletal muscle reveals that insulin regulates approximately 800 genes during a hyperinsulinemic clamp. J Biol Chem 2003, 278:18063–18068. Romero J, Gerber C: Levator scapulae and rhomboid transfer for paralysis of trapezius - The Eden-Lange procedure. J Bone Joint Surg (Br) 2003, 85B:1141–1145. Rosenson RS: Current overview of statin-induced myopathy. Am J Med 2004, 116:408–416. Rothfork JM, DessusBabus S, VanWamel WJB, et al.: Fibrinogen depletion attenuates Staphyloccocus aureus infection by preventing densitydependent virulence gene UP-regulation. J Immunol 2003, 171:5389–5395. Russo S, Bussolati B, Deambrosis I, et al.: Plateletactivating factor mediates CD40-dependent angiogenesis and endothelial-smooth muscle cell interaction. J Immunol 2003, 171:5489– 5497. Rutgers A, Damoiseaux J, Roozendaal C, et al.: ANCA-GBM dot-blot: Evaluation of an assay in the differential diagnosis of patients presenting with rapidly progressive glomerulonephritis. J Clin Immunol 2004, 24:435–440. Rutgers A, Heeringa P, Tervaert JWC: The role of myeloperoxidase in the pathogenesis of systemic vasculitis. Clin Exp Rheumatol 2003, 21:S55–S63. Sabbatini A, Tacchetti C, Tommasi S, et al.: Use of human chondrocyte cell cultures to identify and characterize reactive antibodies in rheumatoid arthritis sera. Clin Exp Rheumatol 2003, 21:587–592. Saeki T, Fujita N, Kourakata H, et al.: Two cases of hypertrophic pachymeningitis associated with myeloperoxidase antineutrophil cytoplasmic autoantibody (MPO-ANCA)-positive pulmonary silicosis in tunnel workers. Clin Rheumatol 2004, 23:76–80. Sahashi K, Ibi T, Ohno K, et al.: Progressive myopathy with circulating autoantibody against giantin in the Golgi apparatus. Neurology 2004, 62:1891–1893. Saino H, Luther F, Carter DH, et al.: Evidence for an extensive collagen Type III proximal domain in the rat femur II. Expansion with exercise. Bone 2003, 32:660–668. Salem GJ, Salinas R, Harding FV: Bilateral kinematic and kinetic analysis of the squat exercise after anterior cruciate ligament reconstruction. Arch Phys Med Rehabil 2003, 84:1211–1216. Salemi S, Aeschlimann A, Gay RE, et al.: Expression and localization of opioid receptors in muscle satellite cells: no difference between fibromyalgia patients and healthy subjects. Arthritis Rheum 2003, 48:3291–3293. Salomonsson S, Ottosson L, Safsten P, et al.: Cloning and characterization of two human Ro52specific monoclonal autoantibodies directed Myositis and myopathies: Miscellaneous 765 towards a domain associated with congenital heart block. J Autoimmun 2004, 22:167–177. Samimi SS, Lesley WS: Craniocervical CT and MR imaging of Schwartz-Jampel syndrome. AJNR Am J Neuroradiol 2003, 24:1694–1696. Sanchez D, Tuckova L, Mothes T, et al.: Epitopes of calreticulin recognised by IgA autoantibodies from patients with hepatic and coeliac disease. J Autoimmun 2003, 21:383–392. Sandler NG, MentinkKane MM, Cheever AW, et al.: Global gene expression profiles during acute pathogen-induced pulmonary inflammation reveal divergent roles for Th1 and Th2 responses in tissue repair. J Immunol 2003, 171:3655–3667. Sato N, Katsumata N, Kagami M, et al.: Clinical assessment and mutation analysis of Kallmann syndrome 1 (KAL1) and fibroblast growth factor receptor 1 (FGFR1, or KAL2) in five families and 18 sporadic patients. J Clin Endocrinol Metab 2004, 89:1079–1088. Sato H, Lagan AL, Alexopoulou C, et al.: The TNF863A allele strongly associates with anticentromere antibody positivity in scleroderma. Arthritis Rheum 2004, 50:558–564. Satoh M, Kuroda Y, Yoshida H, et al.: Induction of lupus autoantibodies by adjuvants. J Autoimmun 2003, 21:1–9. Saxena S, Madan T, Muralidhar K, et al.: cDNA cloning, expression and characterization of an allergenic L3 ribosomal protein of Aspergillus fumigatus. Clin Exp Immunol 2003, 134:86– 91. Schmidt E, Wehr B, Tabengwa EM, et al.: Elevated expression and release of tissue-type, but not urokinase-type, plasminogen activator after binding of autoantibodies to bullous pemphigoid antigen 180 in cultured human keratinocytes. Clin Exp Immunol 2004, 135:497–504. Schmidt J, Rakocevic G, Raju R, et al.: Upregulated inducible co-stimulator (ICOS) and ICOS-ligand in inclusion body myositis muscle: significance for CD8(+) T cell cytotoxicity. Brain 2004, 127:1182–1190. Schmitter T, Agerer F, Peterson L, et al.: Granulocyte CEACAM3 is a phagocytic receptor of the innate immune system that mediates recognition and elimination of human-specific pathogens. J Exp Med 2004, 199:35–46. Schnappinger D, Ehrt S, Voskuil MI, et al.: Transcriptional adaptation of Mycobacterium tuberculosis within macrophages: Insights into the phagosomal environment. J Exp Med 2003, 198:693–704. Schorlemmer S, Gohl C, Iwabu S, et al.: Glucocorticoid treatment of ovariectomized sheep affects mineral density, structure, and mechanical properties of cancellous bone. J Bone Miner Res 2003, 18:2010–2015. Schultz H, Heintz H, vanZandbergen G, et al.: ANCA against the bactericidal/permeability increasing protein (BPI-ANCA) can compromise the antibiotic function of BPI in a Wegener’s granulomatosis patient. Clin Exp Rheumatol 2003, 21:763–766. Schumacher HR, Pessler F, Chen LX: Diagnosing early rheumatoid arthritis (RA). What are the problems and opportunities? Clin Exp Rheumatol 2003, 21:S15–S19. Schwarze C, Terjung B, Lilienweiss P, et al.: IgA class antineutrophil cytoplasmic antibodies in primary sclerosing cholangitis and autoimmune hepatitis. Clin Exp Immunol 2003, 133:283– 289. Seelen MA, Trouw LA, VanderHoorn JWA, et al.: Autoantibodies against mannose-binding lectin in systemic lupus erythematosus. Clin Exp Immunol 2003, 134:335–343. Selcen D, Engel AG: Mutations in myotilin cause myofibrillar myopathy. Neurology 2004, 62: 1363–1371. Selga D, Segelmark M, Wieslander J, et al.: Epitope mapping of anti-PR3 antibodies using chimeric human/mouse PR3 recombinant proteins. Clin Exp Immunol 2004, 135:164–172. Shahrara S, Amin MA, Woods JM, et al.: Chemokine receptor expression and in vivo signaling pathways in the joints of rats with adjuvantinduced arthritis. Arthritis Rheum 2003, 48: 3568–3583. Sharif MN, Tassiulas I, Hu Y, et al.: IFN-alpha priming results in a gain of proinflammatory function by IL-10: Implications for systemic lupus erythematosus pathogenesis. J Immunol 2004, 172:6476–6481. Shaw T, Quan J, Totoritis MC: B cell therapy for rheumatoid arthritis: the rituximab (Anti-CD20) experience. Ann Rheum Dis 2003, 62:55–59. Shen CR, Youssef AR, Devine A, et al.: Peptides containing a dominant T-cell epitope from red cell band 3 have in vivo immunomodulatory properties in NZB mice with autoimmune hemolytic anemia. Blood 2003, 102:3800–3806. Silva M, Shepherd EF, Jackson WO, et al.: Knee strength after total knee arthroplasty. J Arthroplasty 2003, 18:605–611. Silva EEC, Ramos OP, Bauab TM, et al.: Yersinia enterocolitica O:3 isolated from patients with or without reactive arthritis induces polyclonal activation of B cells and autoantibody production in vivo. Autoimmunity 2003, 36:261–268. Silverman GJ, Weisman S: Rituximab therapy and autoimmune disorders: prospeccts for anti-B cell therapy. Arthritis Rheum 2003, 48:1484– 1492. Silverman GJ, Carson DA: Roles of B cells in rheumatoid arthritis. Arthritis Res Ther 2003, 5:S1–S6. Silvestri G, Feinberg MB: Turnover of lymphocytes and conceptual paradigms in HIV infection. J Clin Invest 2003, 112:821–824. Simelyte E, Rimpilainen M, Zhang X, et al.: Role of peptidoglycan subtypes in the pathogenesis of bacterial cell wall arthritis. Ann Rheum Dis 2003, 62:976–982. Singleton A, Myers A, Hardy J: The law of mass action applied to neurodegenerative disease: a hypothesis concerning the etiology and pathogenesis of complex diseases. Hum Mol Genet 2004, 13:R123–R126. SipekDolnicar A, Hojnik M, Rozman B: Bleeding tendency and relapsing small vessel thromboses in a patient with secondary antiphospholipid syndrome. Clin Exp Rheumatol 2003, 21: 412–413. Sjogren H, Meis-Kindblom JM, Orndal C, et al.: Studies on the molecular pathogenesis of extraskeletal myxoid chondrosarcoma cytogenetic, molecular genetic, and cDNA microarray analyses. Am J Pathol 2003, 162:781–792. Sjowall C, Bengtsson AA, Sturfelt G, et al.: Serum levels of autoantibodies against monomeric C-reactive protein are correlated with disease activity in systemic lupus erythematosus. Arthritis Res Ther 2004, 6:R87–R94. Skoldberg F, Rorsman F, Perheentupa J, et al.: Analysis of antibody reactivity against cysteine sulfinic acid decarboxylase, a pyridoxal phosphate-dependent enzyme, in endocrine autoimmune disease. J Clin Endocrinol Metab 2004, 89:1636–1640. Smeets TJM, Barg EC, Kraan MC, et al.: Analysis of the cell infiltrate and expression of proinflammatory cytokines and matrix metalloproteinases in arthroscopic synovial biopsies: comparison with synovial samples from patients with end stage, destructive rheumatoid arthritis. Ann Rheum Dis 2003, 62:635–638. Smeets B, TeLoeke NAJM, Dijkman HBPM, et al.: The parietal epithelial cell: A key player in the pathogenesis of focal segmental glomerulosclerosis in Thy-1.1 transgenic mice. J Am Soc Nephrol 2004, 15:928–939. Smith J, Malanga GA, Yu B, et al.: Effects of functional knee bracing on muscle-firing patterns about the chronic anterior cruciate ligamentdeficient knee. Arch Phys Med Rehabil 2003, 84:1680–1686. Smith AG, Singleton JR: The diagnostic yield of a standardized approach to idiopathic sensorypredominant neuropathy. Arch Intern Med 2004, 164:1021–1025. Smith TJ: The putative role of fibroblasts in the pathogenesis of Graves’ disease: Evidence for the involvement of the insulin-like growth factor-1 receptor in fibroblast activation. Autoimmunity 2003, 36:409–415. Soderbergh A, Myhre AG, Ekwall O, et al.: Prevalence and clinical associations of 10 defined autoantibodies in autoimmune polyendocrine syndrome type I. J Clin Endocrinol Metab 2004, 89:557–562. Sokka T, Willoughby J, Yazici Y, et al.: Databases of patients with early rheumatoid arthritis in the USA. Clin Exp Rheumatol 2003, 21:S146– S153. Sola B, Troussard X: Relevance of cyclin D1 level in the pathogenesis of multiple myeloma. Blood 2003, 102:4245–4246. Soverini S, Martinelli G, Cavo M: Cyclin D1 expression in multiple myeloma: a pathogenetic role besides the prognostic relevance? - Response. Blood 2003, 102:4246. Staikou EV, Routsias JG, Makri AA, et al.: Calreticulin binds preferentially with B cell linear epitopes of Ro60 kD autoantigen, enhancing recognition by anti-Ro60 kD autoantibodies. Clin Exp Immunol 2003, 134:143–150. Staton CA, Brown NJ, Rodgers GR, et al.: Alphastatin, a 24-amino acid fragment of human fibrinogen, is a potent new inhibitor of activated endothelial cells in vitro and in vivo. Blood 2004, 103:601–606. Steidle C, Schwartz S, Jacoby K, et al.: AA2500 testosterone gel normalizes androgen levels in aging males with improvements in body composition and sexual function. J Clin Endocrinol Metab 2003, 88:2673–2681. Steptoe RJ, Stankovic S, Lopaticki S, et al.: Persistence of recipient lymphocytes in NOD mice after irradiation and bone marrow transplantation. J Autoimmun 2004, 22:131–138. Straub RH, Scholmerich J, Cutolo M: The multiple facets of premature aging in rheumatoid arthritis. Arthritis Rheum 2003, 48:2713–2721. Strombeck B, Ekdahl C, Manthorpe R, et al.: Physical capacity in women with primary Sjogren’s syndrome: A controlled study. Arthritis Rheum Arthritis Care Res 2003, 49:681–688. Studt JD, Hovinga JAK, Radonic R, et al.: Familial acquired thrombotic thrombocytopenic purpura: ADAMTS13 inhibitory autoantibodies in identical twins. Blood 2004, 103:4195–4197. Sung DH, Jung JY, Kim HD, et al.: Motor branch of the rectus femoris: Anatomic location for selective motor branch block in stiff-legged gait. Arch Phys Med Rehabil 2003, 84:1028–1031. Suzue K, Asai T, Takeuchi T, et al.: In vivo role of IFN-gamma produced by antigen-presenting cells in early host defense against intracellular pathogens. Eur J Immunol 2003, 33:2666– 2675. Szepesi K, Posan E, Harsfalvi J, et al.: The most severe forms of Perthes’ disease associated with the homozygous Factor V Leiden mutation. J Bone Joint Surg (Br) 2004, 86B:426– 429. Taivassalo T, Jensen TD, Kennaway N, et al.: The spectrum of exercise tolerance in mitochondrial myopathies - A study of 40 patients. Brain 2003, 126:413–423. Takagi K, Kawaguchi Y, Hara M, et al.: Serum nitric oxide (NO) levels in systemic sclerosis patients: correlation between NO levels and clinical features. Clin Exp Immunol 2003, 134: 538–544. Takahashi R, Tsutsumi A, Ohtani K, et al.: Anti-mannose binding lectin antibodies in sera of Japanese patients with systemic lupus erythematosus. Clin Exp Immunol 2004, 136:585–590. Takeda S, Kondo M, Sasaki J, et al.: Fukutin is required for maintenance of muscle integrity, 766 Myositis and myopathies: Miscellaneous cortical histiogenesis and normal eye development. Hum Mol Genet 2003, 12:1449–1459. Tan FK, Tercero GM, Arnett FC, et al.: Examination of the possible role of biologically relevant genes around FBN1 in systemic sclerosis in the Choctaw population. Arthritis Rheum 2003, 48:3295–3296. Tan EM, Shi FD: Relative paradigms between autoantibodies in lupus and autoantibodies in cancer. Clin Exp Immunol 2003, 134:169–177. Temperley RJ, Seneca SH, Tonska K, et al.: Investigation of a pathogenic mtDNA microdeletion reveals a translation-dependent deadenylation decay pathway in human mitochondria. Hum Mol Genet 2003, 12:2341–2348. Tenschert S, Reinert A, Hoheisel U, et al.: Effects of a chronic myositis on structural and functional features of spinal astrocytes in the rat. Neurosci Lett 2004, 361:196–199. Terzoglou AG, Routsias JG, Sakarellos C, et al.: Linear epitopes of two different autoantigensLa/SSB and myelin basic protein - with a high degree of molecular similarity, cause different humoral immune responses. J Autoimmun 2003, 21:47–57. Thakurdas SM, Hasan Z, Hussain R: IgG1 antimycobacterial antibodies can reverse the inhibitory effect of pentoxifylline on tumour necrosis factor alpha (TNF-alpha) secreted by mycobacterial antigen-stimulated adherent cells. Clin Exp Immunol 2004, 136:320–327. Thomzig A, SchulzSchaeffer W, Kratzel C, et al.: Preclinical deposition of pathological prion protein PrPSc in muscles of hamsters orally exposed to scrapie. J Clin Invest 2004, 113: 1465–1472. Thrasyvoulides A, Lymberi P: Evidence for intramolecular B-cell epitope spreading during experimental immunization with an immunogenic thyroglobulin peptide. Clin Exp Immunol 2003, 132:401–407. Tobian AAR, Potter NS, Ramachandra L, et al.: Alternate class I MHC antigen processing is inhibited by toll-like receptor signaling pathogenassociated molecular patterns: Mycobacterium tuberculosis 19-kDa lipoprotein, CpG DNA, and lipopolysaccharide. J Immunol 2003, 171: 1413–1422. Tomimitsu H, Shimizu J, Ishikawa K, et al.: Distal myopathy with rimmed vacuoles (DMRV) New GNE mutations and splice variant. Neurology 2004, 62:1607–1610. Torn C, Gupta M, Zake LN, et al.: Heterozygosity for MICA5.0/MICA5.1 and HLA-DR3DQ2/DR4-DQ8 are independent genetic risk factors for latent autoimmune diabetes in adults. Hum Immunol 2003, 64:902–909. Tran HB, Ohlsson M, Beroukas D, et al.: Subcellular redistribution of La(SS-B) autoantigen during physiologic apoptosis in the fetal mouse heart and conduction system: a clue to the pathogenesis of congenital heart block. Arthritis Rheum 2002, 46:202–208. Trautner BW, Darouiche RO: Catheter-associated infections - Pathogenesis affects prevention. Arch Intern Med 2004, 164:842–850. Tripathy NK, Sinha N, Nityanand S: Anti-annexin V antibodies in Takayasu’s arteritis: prevalence and relationship with disease activity. Clin Exp Immunol 2003, 134:360–364. Trouw LA, Seelen MA, Visseren R, et al.: Anti-C1q autoantibodies in murine lupus nephritis. Clin Exp Immunol 2004, 135:41–48. Trudel G, Jabi M, Uhthoff HK: Localized and adaptive synoviocyte proliferation characteristics in rat knee joint contractures secondary to immobility. Arch Phys Med Rehabil 2003, 84:1350– 1356. Tsai NT, McClure PW, Karduna AR: Effects of muscle fatigue on 3-dimensional scapular kinematics. Arch Phys Med Rehabil 2003, 84:1000–1005. Tsuruta Y, Ikezoe K, Nakagaki H, et al.: A case of dermato-fasciitis: amyopathic dermatomyositis associated with fasciitis. Clin Rheumatol 2004, 23:160–162. Tugay M, Yildiz F, Utkan T, et al.: Esophagitis impairs esophageal smooth muscle reactivity in the rat model: An in vitro study. Dig Dis Sci 2003, 48:2147–2152. Tunca M, BenChetrit E: Familial Mediterranean fever in 2003. Pathogenesis and management. Clin Exp Rheumatol 2003, 21:S49–S52. Tzioufas AG, Wassmuth R, Dafni UG, et al.: Clinical, immunological, and immunogenetic aspects of autoantibody production against SSA/Ro, SSB/La and their linear epitopes in primary Sjogren’s syndrome (pSS): a European multicentre study. Ann Rheum Dis 2002, 61:398–404. Uchida K, Nakata K, Trapnell BC, et al.: High-affinity autoantibodies specifically eliminate granulocyte-macrophage colony-stimulating factor activity in the lungs of patients with idiopathic pulmonary alveolar proteinosis. Blood 2004, 103:1089–1098. Ulanet DB, Wigley FM, Gelber AC, et al.: Autoantibodies against B23, a nucleolar phosphoprotein, occur in scleroderma and are associated with pulmonary hypertension. Arthritis Rheum 2003, 49:85–92. Ulanet DB, Flavahan NA, CasciolaRosen L, et al.: Selective cleavage of nucleolar autoantigen B23 by granzyme B in differentiated vascular smooth muscle cells - Insights into the association of specific autoantibodies with distinct disease phenotypes. Arthritis Rheum 2004, 50:233–241. UusiRasi K, Beck TJ, Sievanen H, et al.: Associations of hormone replacement therapy with bone structure and physical performance among postmenopausal women. Bone 2003, 32:704–710. Valianpour : Cardiolipin deficiency in X-linked cardioskeletal myopathy and neutropenia (Barth syndrome, MIM 302060): A study in cultured skin fibroblasts (Vol 141, pg 729, 2002). J Pediatr 2004, 144:142. Varadi K, Schreiner J, Plaimauer B, et al.: ADAMTS13 autoantibody detection by quantitative immunoblotting. Blood 2003, 102:1932– 1933. Vattemi G, Tonin P, Mora M, et al.: Expression of protein kinase C isoforms and interleukin-1 beta in myofibrillar myopathy. Neurology 2004, 62:1778–1782. Viskari H, Paronen J, Keskinen P, et al.: Humoral beta-cell autoimmunity is rare in patients with the congenital rubella syndrome. Clin Exp Immunol 2003, 133:378–383. Visser M, Deeg DJH, Lips P: Low vitamin D and high parathyroid hormone levels as determinants of loss of muscle strength and muscle mass (Sarcopenia): The Longitudinal Aging Study Amsterdam. J Clin Endocrinol Metab 2003, 88:5766–5772. Vittecoq O, Incaurgarat B, JouenBeades F, et al.: Autoantibodies recognizing citrullinated rat filaggrin in an ELISA using citrullinated and non-citrullinated recombinant proteins as antigens are highly diagnostic for rheumatoid arthritis. Clin Exp Immunol 2004, 135:173–180. Vlieland TPMV: Rehabilitation of people with rheumatoid arthritis. Best Pract Res Clin Rheumatol 2003, 17:847–861. Vossenaar ER, Zendman AJW, vanVenrooij WJ: Citrullination, a possible functional link between susceptibility genes and rheumatoid arthritis. Arthritis Res Ther 2003, 6:1–5. Wake DJ, Rask E, Livingstone DEW, et al.: Local and systemic impact of transcriptional upregulation of 11 beta-hydroxysteroid dehydrogenase type 1 in adipose tissue in human obesity. J Clin Endocrinol Metab 2003, 88:3983– 3988. Wang Y, Zhao MH, Zhang YK, et al.: Binding capacity and pathophysiological effects of IgA1 from patients with IgA nephropathy on human glomerular mesangial cells. Clin Exp Immunol 2004, 136:168–175. Wang HS, Clarke SH: Regulation of B-cell development by antibody specificity. Curr Opin Immunol 2004, 16:246–250. Wang W, Ostlie NS, ContiFine BM, et al.: The susceptibility to experimental myasthenia gravis of STAT(-/-) and STAT(-/-) BALB/c mice suggests a pathogenic role of Th1 cells. J Immunol 2004, 172:97–103. Watanabe A, Kodera M, Sugiura K, et al.: AntiDFS70 antibodies in 597 healthy hospital workers. Arthritis Rheum 2004, 50:892–900. Waters ST, McDuffie M, Bagavant H, et al.: Breaking tolerance to double stranded DNA, nucleosome, and other nuclear antigens is not required for the pathogenesis of lupus glomerulonephritis. J Exp Med 2004, 199:255–264. Weber GF: The absence of CD44 ameliorates Fas(Lpr/lpr) disease. Autoimmunity 2004, 37:1–8. Westervelt P, Lane AA, Pollock JL, et al.: High-penetrance mouse model of acute promyelocytic leukemia with very low levels of PML-RAR alpha expression. Blood 2003, 102:1857–1865. Weston MD, Luijendijk MWJ, Humphrey KD, et al.: Mutations in the VLGR1 gene implicate Gprotein signaling in the pathogenesis of Usher syndrome type II. Am J Hum Genet 2004, 74: 357–366. Weston : Mutations in the VLGR1 gene implicate G-protein signaling in the pathogenesis of Usher syndrome type II (Vol 74, pg 357, 2003). Am J Hum Genet 2004, 74:1080. Whyburn LR, Halcomb KE, Contreras CM, et al.: Reduced dosage of Bruton’s tyrosine kinase uncouples B cell hyperresponsiveness from autoimmunity in lyn(-/-) mice. J Immunol 2003, 171:1850–1858. Wiesner TD, Bluher M, Windgassen M, et al.: Improvement of insulin sensitivity after adrenalectomy in patients with pheochromocytoma. J Clin Endocrinol Metab 2003, 88:3632–3636. Willison HJ, Townson K, Veitch J, et al.: Synthetic disialylgalactose immunoadsorbents deplete anti-GQ1b antibodies from autoimmune nueropathy sera. Brain 2004, 127:680–691. Wilson SG, Reed PW, Andrew T, et al.: A genomescreen of a large twin cohort reveals linkage for quantitative ultrasound of the calcaneus to 2q33-37 and 4q12-21. J Bone Miner Res 2004, 19:270–277. Wilson A, HsingTing Y, Goodnough LT, et al.: Prevalence and outcomes of anemia in rheumatoid arthritis: A systematic review of the literature. Am J Med 2004, 116:50–57. Wolff J, Brendel C, Fink L, et al.: Lack of NB1 GP (CD177/HNA-2a) gene transcription in NB1 GP(−) neutrophils from NB1 GP-expressing individuals and association of low expression with NB1 gene polymorphisms. Blood 2003, 102:731–733. Worda M, Sgonc R, Dietrich H, et al.: In vivo analysis of the apoptosis-inducing effect of antiendothelial cell antibodies in systemic sclerosis by the chorionallantoic membrane assay. Arthritis Rheum 2003, 48:2605–2614. Wuthrich M, Filutowicz HI, Warner T, et al.: Vaccine immunity to pathogenic fungi overcomes the requirement for CD4 help in exogenous antigen presentation to CD8(+) T cells: Implications for vaccine development in immune-deficient hosts. J Exp Med 2003, 197:1405–1416. Wuthrich M, Filutowicz HI, Warner T, et al.: Vaccine immunity to pathogenic fungi overcomes the requirement for CD4 help in exogenous antigen presentation to CD8+ T cells - Implications for vaccine development in immune-deficient hosts. J Exp Med 2003, 197:1405–1416. Xiang Y, Sekine T, Nakamura H, et al.: Proteornic surveillance of autoimmunity in osteoarthritis - Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Raynaud and vascular manifestations 767 Identification of triosephosphate isomerase as an autoantigen in patients with osteoarthritis. Arthritis Rheum 2004, 50:1511–1521. Yamaguchi A, Katsuyama K, Nagahama K, et al.: Possible role of autoantibodies in the pathophysiology of GM2 gangliosidoses. J Clin Invest 2004, 113:200–208. Yamamoto T: Potential roles of CCL2/monocyte chemoattractant protein-1 in the pathogenesis of cutaneous sclerosis. Clin Exp Rheumatol 2003, 21:369–375. Yan YH, Devos T, Yu LS, et al.: Pathogenesis of autoimmunity after xenogeneic thymus transplantation. J Immunol 2003, 170:5936–5946. Yao L, Salvucci C, Cardones AR, et al.: Selective expression of stromal-derived factor-1 in the capillary vascular endothelium plays a role in Kaposi sarcoma pathogenesis. Blood 2003, 102:3900–3905. Yasunari K, Maeda K, Nakamura M, et al.: Effects of carvedilol on oxidative stress in polymorphonuclear and mononuclear cells in patients with essential hypertension. Am J Med 2004, 116: 460–465. Yasuoka H, Ihn H, Medsger TA, et al.: A novel protein highly expressed in testis is overexpressed in systemic sclerosis fibroblasts and targeted by autoantibodies. J Immunol 2003, 171:6883–6890. Yoshida K, Tsutsumi A, Ohnishi Y, et al.: T cell epitopes of prothrombin in patients with antiphospholipid syndrome. Ann Rheum Dis 2003, 62: 905–906. Yoshimasu T, Nishide T, Seo N, et al.: Susceptibility of T cell receptor-alpha chain knock-out mice to ultraviolet B light and fluorouracil: a novel model for drug-induced cutaneous lupus erythematosus. Clin Exp Immunol 2004, 136: 245–254. Zandbelt MM, Vogelzangs J, vandePutte LBA, et al.: Anti-alpha-fodrin antibodies do not add much to the diagnosis of Sjogren’s syndrome. Arthritis Res Ther 2003, 6:R33–R38. Zhao AP, McDermott J, Urban JF, et al.: Dependence of IL-4, IL-13, and nematode-induced alterations in murine small intestinal smooth muscle contractility on Stat6 and enteric nerves. J Immunol 2003, 171:948–954. Zheng D, Wolfe M, Cowley BD, et al.: Urinary excretion of monocyte chemoattractant protein-1 in autosomal dominant polycystic kidney disease. J Am Soc Nephrol 2003, 14:2588–2595. Zimmermann N, King NE, Laporte J, et al.: Dissection of experimental asthma with DNA microarray analysis identifies arginase in asthma pathogenesis. J Clin Invest 2003, 111:1863– 1874. Zloza A, Sullivan YB, Connick E, et al.: CD8(+) T cells that express CD4 on their surface (CD4(Dim)CD8(Bright) T cells) recognize an antigen-specific target, are detected in vivo, and can be productively infected by T-tropic HIV. Blood 2003, 102:2156–2164. Zobel ATC, Loranger A, Marceau N, et al.: Distinct chaperone mechanisms can delay the formation of aggresomes by the myopathy-causing R120G alpha B-crystallin mutant. Hum Mol Genet 2003, 12:1609–1620. delaMotte CA, Hascall VC, Drazba J, et al.: Mononuclear leukocytes bind to specific hyaluronan structures on colon mucosal smooth muscle cells treated with polyinosinic acid: Polycytidylic acid - Inter-alpha-trypsin inhibitor is crucial to structure and function. Am J Pathol 2003, 163:121–133. van Blokland SC, Versnel MA: Pathogenesis of Sjogren’s syndrome: characteristics of different mouse models for autoimmune exocrinopathy. Clin Immunol 2002, 103:111–124. vanGaalen FA, Toes REM, Ditzel HJ, et al.: Association of autoantibodies to glucose-6-phosphate isomerase with extraarticular complications in rheumatoid arthritis. Arthritis Rheum 2004, 50:395–399. vanLent P, Nabbe KC, Boross P, et al.: The inhibitory receptor Fc gamma RII reduces joint inflammation and destruction in experimental immune complex-mediated arthritides not only by inhibition of Fc gamma RI/III but also by efficient clearance and endocytosis of immune complexes. Am J Pathol 2003, 163:1839– 1848. vanRossum AP, Limburg PC, Kallenberg CGM: Membrane proteinase 3 expression on resting neutrophils as a pathogenic factor in PR3ANCA-associated vasculitis. Clin Exp Rheumatol 2003, 21:S64–S68. vandeVlekkert J, TjinATon MLR, Hoogendijk JE: Quality of myositis case reports open to improvement. Arthritis Rheum Arthritis Care Res 2004, 51:148–150. Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes Raynaud and vascular manifestations: pathophysiology, diagnosis, and management Review (pp 718–722) Anderson JE, Held N, Wright K: Raynaud’s phenomenon of the nipple: A treatable cause of painful breastfeeding. Pediatrics 2004, 113:E360–E364. AnglesCano E: How statins and fibrates lower CRP. Blood 2004, 103:3996–3997. Bredemeier M, Xavier RM, Capobianco KG, et al.: Nailfold capillary microscopy can suggest pulmonary disease activity in systemic sclerosis. J Rheumatol 2004, 31:286–294. Bregenzer N, Distler O, Meyringer R, et al.: Doppler ultrasound identifies increased resistive indices in SSc. Ann Rheum Dis 2004, 63:109– 110. Cerinic MM, Valentini G, Sorano GG, et al.: Blood coagulation, fibrinolysis, and markers of endo• thelial dysfunction in systemic sclerosis. Semin Arthritis Rheum 2003, 32:285–295. [13]. Clowse MEB, Wigley FM: Digital necrosis related to carboplatin and gemcitabine therapy in systemic sclerosis. J Rheumatol 2003, 30:1341– 1343. Cutolo M, Grassi W, Cerinic MM: Raynaud’s phenomenon and the role of capillaroscopy. Arthritis Rheum 2003, 48:3023–3030. DeAngelis R, DelMedico P, Blasetti P, et al.: Raynaud’s Phenomenon: Clinical Spectrum of 118 Patients. Clin Rheumatol 2003, 22:279– 284. DelPapa N, Colombo G, Fracchiolla N, et al.: Circu•• lating endothelial cells as a marker of ongoing vascular disease in systemic sclerosis. Arthritis Rheum 2004, 50:1296–1304. [18]. Economides PA, Caselli A, Tiani E, et al.: The effects of atorvastatin on endothelial function in diabetic patients and subjects at risk for type 2 diabetes. J Clin Endocrinol Metab 2004, 89: 740–747. Fontana L, Marion MJ, Catilina P: Persistent Raynaud’s phenomenon after exposure to vinyl chloride monomer: Assessment of endothelial damage. Clin Exp Rheumatol 2004, 22:132– 133. Furspan PB, Chatterjee S, Freedman RR: Increased tyrosine phosphorylation mediates the coolinginduced contraction and increased vascular reactivity of Raynaud’s disease. Arthritis Rheum 2004, 50:1578–1585. Goligorsky MS, Wang WH: Statins’ coat of many colors receives yet another hue. J Am Soc Nephrol 2004, 15:1098–1100. Jansen TLTA, Janssens PMW: Transient cryofibrinogenaemia with acral digital necrosis, secondary Raynaud’s phenomenon and polyarthritis. Clin Exp Rheumatol 2003, 21:407–408. Johnson S, Iazzetta J, Dewar C: Severe Raynaud’s phenomenon with yohimbine therapy for erectile dysfunction. J Rheumatol 2003, 30:2503– 2505. Jones GT, Herrick AL, Woodham SE, et al.: Occurrence of Raynaud’s phenomenon in children ages 12-15 years - Prevalence and association with other common symptoms. Arthritis Rheum 2003, 48:3518–3521. Kleemann R, Verschuren L, deRooij BJ, et al.: Evidence for anti-inflammatory activity of statins and PPAR alpha activators in human C-reactive protein transgenic mice in vivo and in cultured human hepatocytes in vitro. Blood 2004, 103:4188–4194. Konttinen YT, Mackiewicz Z, Ruuttila P, et al.: Vascular damage and lack of angiogenesis in sys• temic sclerosis skin. Clin Rheumatol 2003, 22:196–202. [17]. Kotsis SV, Chung KC: A systematic review of the outcomes of digital sympathectomy for treatment of chronic digital ischemia. J Rheumatol 2003, 30:1788–1792. Mackay AC: Raynaud’s phenomenon in a pneumatic tool worker (Reprinted from Trans Assoc Ind Med Officers, vol 3, pg 266-267, 1953). Occup Med - Oxford 2003, 53:342. Montisci R, Vacca A, Garau P, et al.: Detection of early impairment of coronary flow reserve in patients with systemic sclerosis. Ann Rheum Dis 2003, 62:890–893. Nigrovic PA, Fuhlbrigge RC, Sundel RP: Raynaud’s phenomenon in children - A retrospective re• view of 123 patients. Pediatrics 2003, 111: 715–721. [2]. Noel B: Autoimmune disease and other potential side-effects of statins. Lancet 2004, 363:2000. Noel B: Risks and benefits of statins in lupus erythematosus. Arch Intern Med 2004, 164:107– 108. Oskay T, Olmez U: Primary Raynaud’s phenomenon in monozygotic twins. Ann Rheum Dis 2004, 63:219. Parekh S, Iyer R, Sequeira W: Transient renal dysfunction with Raynaud’s phenomenon - Is there a connection? JCR - J Clin Rheumatol 2003, 9:88–91. Porciello G, Scarpato R, Ferri C, et al.: Spontaneous chromosome damage (Micronuclei) in systemic sclerosis and Raynaud’s phenomenon. J Rheumatol 2003, 30:1244–1247. Rajagopalan S, Pfenninger D, Kehrer C, et al.: Increased asymmetric dimethylarginine and endothelin 1 levels in secondary Raynaud’s phenomenon - Implications for vascular dysfunction and progression of disease. Arthritis Rheum 2003, 48:1992–2000. Sulli A, Ghio M, Bezante GP, et al.: Blunted coronary flow reserve in systemic sclerosis. Rheumatology 2004, 43:505–509. Wallace DJ: Historical vignette: Raynaud’s phenomenon as perceived by musculoskeletal specialists (1862-1957). JCR - J Clin Rheumatol 2003, 9:87. Whitfield ML, Finlay DR, Murray JI, et al.: Systemic •• and cell type-specific gene expression patterns in scleroderma skin. Proc Natl Acad Sci USA 2003, 100:12319–12324. [19]. Worda M, Sgonc R, Dietrich H, et al.: In vivo analy•• sis of the apoptosis-inducing effect of antiendothelial cell antibodies in systemic sclerosis by the chorionallantoic membrane assay. Arthritis Rheum 2003, 48:2605–2614. [15]. Yamashita M, Funayama H, Anami Y, et al.: Cyanotic change of colon mucosa in a patient with Raynaud’s phenomenon. Am J Med 2004, 116:570–572. Ziegler S, Brunner M, Eigenbauer E, et al.: Longterm outcome of primary Raynaud’s phenom• enon and its conversion to connective tissue 768 Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Autoantibodies: pathogenetic significance, clinical utility disease - A 12-year retrospective patient analysis. Scand J Rheumatol 2003, 32:343– 347. [4]. Ziegler S, Brunner M, Eigenbauer E, et al.: Longterm outcome of primary Raynaud’s phenomenon and its conversion to connective tissue disease: a 12-year retrospective patient analysis. Scand J Rheumatol 2003, 32:346–350. vanVugt RM, Kater L, Dijkstral PF, et al.: The outcome of angiography in patients with Raynaud’s phenomenon: An unexpected role for atherosclerosis and hypercholesterolemia. Clin Exp Rheumatol 2003, 21:445–450. Autoantibodies: pathogenetic significance, clinical utility Review (pp 723–732) Allcock RJ, Forrest I, Corris PA, et al.: A study of the prevalence of systemic sclerosis in north• east England. Rheumatology 2004, 43:596– 602. [22]. Asherson RA: An unusually complicated case of primary Sjogren’s syndrome: Development of transient ”Lupus-type” autoantibodies following silicone implant rejection. J Rheumatol 2004, 31:196–197. Aslangul E, Ranque B, Papo T: Pseudotumoral retroperitoneal fibrosis and localized vasculitis with very high serum levels of anti-PR3 ANCA. Am J Med 2003, 115:250–252. Audrain MAP, Colonna F, Morio F, et al.: Comparison of different kits in the detection of autoantibodies to cardiolipin and beta2glycoprotein 1. Rheumatology 2004, 43:181–185. Bardoni A, Rossi P, Salvini R, et al.: Autoantibodies to RNA-polymerases in Italian patients with • systemic sclerosis. Clin Exp Rheumatol 2003, 21:301–306. [61]. Bizzaro N, Wiik A: Appropriateness in anti-nuclear antibody testing: From clinical request to strategic laboratory practice. Clin Exp Rheumatol 2004, 22:349–355. Buyon JP, Clancy RM: From antibody insult to fibrosis in neonatal lupus the heart of the matter. Arthritis Res Ther 2003, 5:266–270. Caccavo D, DelPorto F, Garzia P, et al.: Raynaud’s phenomenon and antiphospholipid antibodies in systemic lupus erythematosus: is there an association? Ann Rheum Dis 2003, 62:1003– 1005. Chauhan R, Handa R, Das TP, et al.: Over-expression of TATA binding protein (TBP) and p53 and autoantibodies to these antigens are features of systemic sclerosis, systemic lupus erythematosus and overlap syndromes. Clin Exp Immunol 2004, 136:574–584. Cimaz R, Spence DL, Hornberger L, et al.: Incidence and spectrum of neonatal lupus erythematosus: A prospective study of infants born to mothers with anti-Ro autoantibodies. J Pediatr 2003, 142:678–683. Cline AM, Radic MZ: Murine lupus autoantibodies identify distinct subsets of apoptotic bodies. Autoimmunity 2004, 37:85–93. Dahle C, Skogh T, Aberg AK, et al.: Methods of choice for diagnostic antinuclear antibody (ANA) screening benefit of adding antigenspecific assays to immunofluorescence microscopy. J Autoimmun 2004, 22:241–248. Derk CT, Sakkas LI, Rasheed M, et al.: Autoantibodies in patients with systemic sclerosis and • cancer - A case-control study. J Rheumatol 2003, 30:1994–1996. [33]. Derk CT, Sakkas LI, Rasheed M, et al.: Autoantibodies in patients with systemic sclerosis and cancer: A case-control study. J Rheumatol 2003, 30:1994–1996. FeghaliBostwick C, Medsger TA, Wright TM: Analysis of systemic sclerosis in twins reveals low • concordance for disease and high concordance for the presence of antinuclear antibodies. Arthritis Rheum 2003, 48:1956–1963. [1]. Fernvik EC, Ketelhuth DFJ, Russo M, et al.: The autoantibody repertoire against copper- or macrophage-modified LDL differs in normolipidemics and hypercholesterolemic patients. J Clin Immunol 2004, 24:170–176. Ganapathy V, Casiano CA: Autoimmunity to the nuclear autoantigen DFS70 (LEDGF): What exactly are the autoantibodies trying to tell us? Arthritis Rheum 2004, 50:684–688. Gerli R, Caponi L: Clinical and serological associations of ribosomal P autoantibodies in systemic lupus erythematosus: prospective evaluation in a large cohort of Italian patients: reply. Rheumatology 2003, 42:U11–U12. Guth A, Smith D, Wysocki LJ: Intricacies of antiDNA autoantibodies - The authors respond. J Immunol 2004, 172:3367–3368. Hayakawa I, Sato S, Hasegawa M, et al.: A case of scleroderma spectrum disorder with anticentriole antibody and pulmonary hypertension. Clin Rheumatol 2004, 23:266–268. Hayakawa T, Hasegawa M, Takehara K, et al.: AntiDNA topoisomerase II alpha Autoantibodies in localized scleroderma. Arthritis Rheum 2004, 50:227–232. Hu PQ, Fertig N, Medsger TA, et al.: Correlation of serum anti-DNA topoisomerase I antibody lev• els with disease severity and activity in systemic sclerosis. Arthritis Rheum 2003, 48: 1363–1373. [44]. Innocencio RM, Romaldini JH, Ward LS: High prevalence of thyroid autoantibodies in systemic sclerosis and rheumatoid arthritis but not in the antiphospholipid syndrome. Clin Rheumatol 2003, 22:494. Jankovic M, Casellas R, Yannoutsos N, et al.: RAGs and regulation of autoantibodies. Annu Rev Immunol 2004, 22:485–501. Jeoung D, Lim Y, Lee EB, et al.: Identification of autoantibody against poly (ADP-ribose) polymerase (PARP) fragment as a serological marker in systemic lupus erythematosus. J Autoimmun 2004, 22:87–94. Jolly M, Smaron M, Utest TO, et al.: Are isolated antinucleolar antibodies a marker of scleroderma? JCR - J Clin Rheumatol 2003, 9:291– 295. Kao AH, Lacomis D, Lucas M, et al.: Anti-signal recognition particle autoantibody in patients with and patients without idiopathic inflammatory myopathy. Arthritis Rheum 2004, 50:209– 215. Katsumata Y, Suzuki T, Kuwana M, et al.: Anti-c-Mpl (Thrombopoietin receptor) autoantibody-induced amegakaryocytic thrombocytopenia in a patient with systemic sclerosis. Arthritis Rheum 2003, 48:1647–1651. Khanna D, Aggarwal A, Bhakuni DS, et al.: Bactericidal /permeability-increasing protein and cathepsin G are the major antigenic targets of Antineutrophil cytoplasmic autoantibodies in systemic sclerosis. J Rheumatol 2003, 30: 1248–1252. Kuwana M, Pandey JP, Silver RM, et al.: HLA class II alleles in systemic sclerosis patients with • anti-RNA polymerase I/III antibody - Associations with subunit reactivities. J Rheumatol 2003, 30:2392–2397. [64]. Kuwana M, Pandey JP, Silver RM, et al.: HLA class II alleles in systemic sclerosis patients with anti-RNA polymerase I/III antibody: Associations with subunit reactivities. J Rheumatol 2003, 30:2392–2397. Ma CH, Hui J, Tang JTY, et al.: Antibodies to guanosine triphosphate misidentified as antidouble-stranded DNA antibodies in a patient with antinuclear antibody-negative lupus, due to buckling of insolubilized assay DNA. Arthritis Rheum 2004, 50:1533–1538. Matsudaira R, Takeuchi K, Takasaki Y, et al.: Relationships between autoantibody responses to deletion mutants of Ki antigen and clinical manifestations of lupus. J Rheumatol 2003, 30:1208–1214. Matsushita M, Takasaki Y, Takeuchi K, et al.: Autoimmune response to proteasome activator 28 alpha in patients with connective tissue diseases. J Rheumatol 2004, 31:252–259. Mayes MD, Lacey JV, BeebeDimmer J, et al.: Prevalence, incidence, survival, and disease charac• teristics of systemic sclerosis in a large US population. Arthritis Rheum 2003, 48:2246– 2255. [13]. Momot T, Koch S, Hunzelmann N, et al.: Association of killer cell immunoglobulin-like receptors with scleroderma. Arthritis Rheum 2004, 50: 1561–1565. Radic MZ, Weigert M: Intricacies of anti-DNA autoantibodies. J Immunol 2004, 172:3367. Raijmakers R, Renz M, Wiemann C, et al.: PM-Scl75 is the main autoantigen in patients with the • polymyositis/scleroderma overlap syndrome. Arthritis Rheum 2004, 50:565–569. [50]. Reveille JD, Solomon DH: American College of •• Rheumatology Ad Hoc Committee on Immunologic Testing Guidelines - Evidence-based guidelines for the use of immunologic tests Anticentromere, Scl-70, and nucleolar antibodies. Arthritis Rheum 2003, 49:399–412. [10]. Riente L, Chimenti D, Pratesi F, et al.: Antibodies to tissue transglutaminase and Saccharomyces cerevisiae in Ankylosing spondylitis and psoriatic arthritis. J Rheumatol 2004, 31:920–924. Ruffatti A, Grypiotis P, Todesco S: Antineutrophil cytoplasmic antibodies in patients with systemic sclerosis - Dr. Ruffatti, et al reply. J Rheumatol 2003, 30:2079–2080. Ruffatti A, Ostuni P, Grypiotis P, et al.: Sensitivity and specificity for primary Sjorgren’s syndrome of IgA and IgG anti-alpha-fodrin antibodies detected by ELISA. J Rheumatol 2004, 31:504–507. Sato S, Fujimoto M, Hasegawa M, et al.: Antiphospholipid antibody in localised scleroderma. Ann Rheum Dis 2003, 62:771–774. Sato H, Lagan AL, Alexopoulou C, et al.: The TNF -863A allele strongly associates with anticen• tromere antibody positivity in scleroderma. Arthritis Rheum 2004, 50:558–564. [37]. Sato H, Lagan AL, Alexopoulou C, et al.: The TNF863A allele strongly associates with anticentromere antibody positivity in scleroderma. Arthritis Rheum 2004, 50:558–564. Scofield RH: Autoantibodies as predictors of disease. Lancet 2004, 363:1544–1546. Soderbergh A, Myhre AG, Ekwall O, et al.: Prevalence and clinical associations of 10 defined autoantibodies in autoimmune polyendocrine syndrome type I. J Clin Endocrinol Metab 2004, 89:557–562. Sulli A, Ghio M, Bezante GP, et al.: Blunted coronary flow reserve in systemic sclerosis. Rheu• matology 2004, 43:505–509. [24]. Tonutti E, Bassetti D, Piazza A, et al.: Diagnostic accuracy of Elisa methods as an alternative screening test to indirect immunofluorescence for the detection of antinuclear antibodies. Evaluation of five commercial kits. Autoimmunity 2004, 37:171–176. Tzioufas AG, Wassmuth R, Dafni UG, et al.: Clinical, immunological, and immunogenetic aspects of autoantibody production against SSA/Ro, SSB/La and their linear epitopes in primary Sjogren’s syndrome (pSS): a European multicentre study. Ann Rheum Dis 2002, 61:398–404. Ulanet DB, Wigley FM, Gelber AC, et al.: Autoantibodies against B23, a nucleolar phosphoprotein, occur in scleroderma and are associated with pulmonary hypertension. Arthritis Rheum 2003, 49:85–92. Ulanet DB, Wigley FM, Gelber AC, et al.: Autoantibodies against B23, a nucleolar phosphoprotein, occur in systemic sclerosis and are associated with pulmonary hypertension. Arthritis Rheum 2003, 49:85–92. Watanabe A, Kodera M, Sugiura K, et al.: AntiDFS70 antibodies in 597 healthy hospital workers. Arthritis Rheum 2004, 50:892–900. Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Myofibroblasts, EMT, cellular plasticity in tissue repair, fibrosis 769 Worda M, Sgonc R, Dietrich H, et al.: In vivo analysis of the apoptosis-inducing effect of antiendothelial cell antibodies in systemic sclerosis by the chorionallantoic membrane assay. Arthritis Rheum 2003, 48:2605–2614. Wusirika R, Ferri C, Marin M, et al.: The assessment of anti-endothelial cell antibodies in scleroder• ma-associated pulmonary fibrosis. A study of indirect immunofluorescent and western blot analysis in 49 patients with scleroderma. Am J Clin Pathol 2003, 120:596–606. [104]. Yasuoka H, Ihn H, Medsger TA, et al.: A novel protein highly expressed in testis is overexpressed in systemic sclerosis fibroblasts and targeted by autoantibodies. J Immunol 2003, 171:6883–6890. Zandbelt MM, Vogelzangs J, vandePutte LBA, et al.: Anti-alpha-fodrin antibodies do not add much to the diagnosis of Sjogren’s syndrome. Arthritis Res Ther 2003, 6:R33–R38. vanGaalen FA, LinnRasker SP, vanVenrooij WJ, et al.: Autoantibodies to cyclic citrullinated peptides predict progression to rheumatoid arthritis in patients with undifferentiated arthritis - A prospective cohort study. Arthritis Rheum 2004, 50:709–715. Myofibroblasts, EMT, cellular plasticity in tissue repair, fibrosis: potential implications for scleroderma Review (pp 733–738) Asano Y, Ihn H, Yamane K, et al.: Impaired Smad7Smurf-mediated negative regulation of TGFbeta signaling in scleroderma fibroblasts. J Clin Invest 2004, 113:253–264. Fireman E, Schwartz Y, Mann A, et al.: Effect of montelukast, a cysteinyl receptor antagonist, on myofibroblasts in interstitial lung disease. J Clin Immunol 2004, 24:418–425. Hashimoto N, Jin H, Liu TJ, et al.: Bone marrow•• derived progenitor cells in pulmonary fibrosis. J Clin Invest 2004, 113:243–252. [29]. Hildebrand KA, Zhang M, vanSnellenberg W, et al.: Myofibroblast numbers are elevated in human elbow capsules after trauma. Clin Orthop 2004, 419:189–197. Kalluri R, Neilson EG: Epithelial-mesenchymal tran•• sition and its implications for fibrosis. J Clin Invest 2003, 112:1776–1784. [7]. Koumas L, Smith TJ, Feldon S, et al.: Thy-1 expression in human fibroblast subsets defines myofibroblastic or lipofibroblastic phenotypes. Am J Pathol 2003, 163:1291–1300. Kuwana M, Okazaki Y, Kodama H, et al.: Human circulating CD14+ monocytes as a source of • progenitors that exhibit mesenchymal cell differentiation. J Leukoc Biol 2003, 74:833–845. [28]. Lambert NC, Erickson TD, Yan Z, et al.: Quantification of maternal microchimerism by HLA-specific real-time polymerase chain reaction Studies of healthy women and women with scleroderma. Arthritis Rheum 2004, 50:906– 914. Li JH, Wang WS, Huang XR, et al.: Advanced glycation end products induce tubular epithelialmyofibroblast transition through the RAGEERK1/2 MAP kinase signaling pathway. Am J Pathol 2004, 164:1389–1397. Liu TJ, Dhanasekaran SM, Jin H, et al.: FIZZ1 stimulation of myofibroblast differentiation. Am J Pathol 2004, 164:1315–1326. Momot T, Koch S, Hunzelmann N, et al.: Association of killer cell immunoglobulin-like receptors with scleroderma. Arthritis Rheum 2004, 50: 1561–1565. Offersen BV, Nielsen BS, HoyerHansen G, et al.: The myofibroblast is the predominant plasminogen activator inhibitor-1-expressing cell type in human breast carcinomas. Am J Pathol 2003, 163:1887–1899. Ozbilgin MK, Inan S: The roles of Transforming growth Factor Type beta(3)(TGF-beta(3)) and mast cells in the pathogenesis of scleroderma. Clin Rheumatol 2003, 22:189–195. Pannu J, Gorehyer E, Yamanaka M, et al.: An increased transforming growth factor beta receptor type I:Type II ratio contributes to elevated collagen protein synthesis that is resistant to inhibition via a kinase-deficient transforming growth factor beta receptor type II in scleroderma. Arthritis Rheum 2004, 50: 1566–1577. Pilling D, Buckley CD, Salmon M, et al.: Inhibition of •• fibrocyte differentiation by serum amyloid P1. J Immunol 2003, 171:5537–5546. [30]. Schmidt M, Sun G, Stacey MA, et al.: Identification of circulating fibrocytes as precursors of bronchial myofibroblasts in asthma. J Immunol 2003, 171:380–389. Shephard P, Martin G, SmolaHess S, et al.: Myofibroblast differentiation is induced in keratinocyte-fibroblast co-cultures and is antagonistically regulated by endogenous transforming growth factor-beta and interleukin-1. Am J Pathol 2004, 164:2055–2066. Wallis DD, Tan FK, Kessler R, et al.: Fibrillin 1 abnormalities in dermal fibroblast cultures from first-degree relatives of patients with systemic sclerosis (Scleroderma). Arthritis Rheum 2004, 50:329–332. Yamane K, Ihn H, Tamaki K: Epidermal growth factor up-regulates expression of. transforming growth factor beta receptor type II in human dermal fibroblasts by phosphoinositide 3-kinase/Akt signaling pathway - Resistance to epidermal growth factor stimulation in scleroderma fibroblasts. Arthritis Rheum 2003, 48: 1652–1666. Yang JW, Dai CS, Liu YH: Hepatocyte growth factor suppresses renal interstitial myofibroblast activation and intercepts Smad signal transduction. Am J Pathol 2003, 163:621–632. Zeisberg M, Bottiglio C, Kumar N, et al.: Bone mor•• phogenic protein-7 inhibits progression of chronic renal fibrosis associated with two genetic mouse models. Am J Physiol Renal Physiol 2003, 285:F1060–F1067. [13]. Zhao Y, Glesne D, Huberman E: A human peripheral blood monocytes-derived subset acts as • pluripotent stem cells. Proc Natl Acad Sci U S A 2003, 100:2426–2431. [22]. Recent advances in fibroblast signaling and biology in scleroderma Review (pp 739–745) Asano Y, Ihn H, Yamane K, et al.: Impaired Smad7•• Smurf-mediated negative regulation of TGFbeta signaling in scleroderma fibroblasts. J Clin Invest 2004, 113:253–264. [21]. Asano Y, Ihn H, Yamane K, et al.: Increased expression levels of integrin alpha v beta 5 on scleroderma fibroblasts. Am J Pathol 2004, 164: 1275–1292. Asano Y, Ihn H, Yamane K, et al.: Increased expres•• sion levels of integrin alphavbeta5 on scleroderma fibroblasts. Am J Pathol 2004, 164: 1275–1292. [63]. Asano Y, Ihn H, Yamane K, et al.: Phosphatidylinositol 3-kinase is involved in alpha 2(I) collagen gene expression in normal and scleroderma fibroblasts. J Immunol 2004, 172:7123–7135. Buhling F, Rocken C, Brasch F, et al.: Pivotal role of cathepsin K in lung fibrosis. Am J Pathol 2004, 164:2203–2216. Chizzolini C, Parel Y, DeLuca C, et al.: Systemic sclerosis Th2 cells inhibit collagen production by dermal fibroblasts via membrane-associated tumor necrosis factor alpha. Arthritis Rheum 2003, 48:2593–2604. DiGuglielmo GM, LeRoy C, Goodfellow AF, et al.: •• Distinct endocytic pathways regulate TGFbeta receptor signalling and turnover. Nat Cell Biol 2003, 5:410–421. [25]. Flanders KC: Smad3 as a mediator of the fibrotic response. Int J Exp Pathol 2004, 85:47–64. • [28]. Fujii T, Mimori T, Kimura N, et al.: Pseudoscleroderma associated with transforming growth factor beta 1-producing advanced gastric carcinoma: comment on the article by Varga. Arthritis Rheum 2003, 48:1766–1767. Fukasawa C, Kawaguchi Y, Harigai M, et al.: Increased CD40 expression in skin fibroblasts from patients with systemic sclerosis (SSc): role of CD40-CD154 in the phenotype of SSc fibroblasts. Eur J Immunol 2003, 33:2792– 2800. Hao ZY, Hampel B, Yagita H, et al.: T cell-specific ablation of Fas leads to Fas ligand-mediated lymphocyte depletion and inflammatory pulmonary fibrosis. J Exp Med 2004, 199:1355– 1365. Holsti MA, Chitnis T, Panzo RJ, et al.: Regulation of postsurgical fibrosis by the programmed death-1 inhibitory pathway. J Immunol 2004, 172:5774–5781. Hu B, Wang S, Zhang Y, et al.: A nuclear target for •• interleukin-1alpha - Interaction with the growth suppressor necdin modulates proliferation and collagen expression. Proc Natl Acad Sci U S A 2003, 100:10008–10013. [72]. Hudson LL, Silver RM, Pandey JR: Ethnic differences in cytotoxic T lymphocyte associated antigen 4 genotype associations with systemic sclerosis. J Rheumatol 2004, 31:85–87. Iwano M, Plieth D, Danoff TM, et al.: Evidence for fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest 2002, 110:341–350. Jimenez SA, Derk CT: Following the molecular pathways toward an understanding of the pathogenesis of systemic sclerosis. Ann Intern Med 2004, 140:37+. Jimenez SA, Derk CT: Following the molecular pathways toward an understanding of the patho• genesis of systemic sclerosis. Ann Intern Med 2004, 140:37–50. [1]. Kao AH, Lacomis D, Lucas M, et al.: Anti-signal recognition particle autoantibody in patients with and patients without idiopathic inflammatory myopathy. Arthritis Rheum 2004, 50:209– 215. Kaufman J, Sime PJ, Phipps RP: Expression of CD154 (CD40 ligand) by human lung fibroblasts: Differential regulation by IFN-gamma and IL-13, and implications for fibrosis. J Immunol 2004, 172:1862–1871. Kawaguchi Y, Takagi K, Hara M, et al.: Angiotensin II in the lesional skin of systemic sclerosis pa• tients contributes to tissue fibrosis via angiotensin II type 1 receptors. Arthritis Rheum 2004, 50:216–226. [66]. Kessel A, Rosner I, Rozenbaum M, et al.: Increased CD8+ T cell apoptosis in scleroderma is associated with low levels of NF-kappa B. J Clin Immunol 2004, 24:30–36. Lan HY, Mu W, Tomita N, et al.: Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 2003, 112:1776–1784. Launay D, Peyrat JP, Verdiere A, et al.: Multiplex reverse transcription polymerase chain reaction assessment of sialyltransferase expression in peripheral blood mononuclear cells in systemic sclerosis. J Rheumatol 2004, 31:88–95. Leask A, Denton CP, Abraham DJ: Insights into the molecular mechanism of chronic fibrosis: The role of connective tissue growth factor in scleroderma. J Invest Dermatol 2004, 122: 1–6. Leask A, Abraham DJ: TGF-beta signaling and the •• fibrotic response. FASEB J 2004, 18:816– 827. [36]. Liu TJ, Dhanasekaran SM, Jin H, et al.: FIZZ1 stimulation of myofibroblast differentiation. Am J Pathol 2004, 164:1315–1326. Mori Y, Chen SJ, Varga J: Expression and regulation of intracellular SMAD signaling in scleroderma skin fibroblasts. Arthritis Rheum 2003, 48: 1964–1978. 770 Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Animal models Mori Y, Chen SJ, Varga J: Expression and regulation •• of intracellular SMAD signalling in scleroderma skin fibroblasts. Arthritis Rheum 2003, 48: 1964–1978. [30]. Olman MA, White KE, Ware LB, et al.: Pulmonary edema fluid from patients with early lung injury stimulates fibroblast proliferation through IL-1 beta-induced IL-6 expression. J Immunol 2004, 172:2668–2677. Pannu J, Gore-Hyer E, Yamanaka M, et al.: An in•• creased transforming growth factor beta receptor type I type II ratio contributes to elevated collagen protein synthesis that is resistant to inhibition via a kinase-deficient transforming growth factor beta receptor type II in scleroderma. Arthritis Rheum 2004, 50: 1566–1577. [19]. Sato M, Muragaki Y, Saika S, et al.: Targeted disruption of TGF-beta 1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J Clin Invest 2003, 112:1486–1494. Shegogue D, Trojanowska M: Mammalian target of •• rapamycin positively regulates collagen type I production via a phosphatidylinositol 3-kinaseindependent pathway. J Biol Chem 2004, 279:23166–23175. [59]. Shephard P, Martin G, SmolaHess S, et al.: Myofibroblast differentiation is induced in keratinocyte-fibroblast co-cultures and is antagonistically regulated by endogenous transforming growth factor-beta and interleukin-1. Am J Pathol 2004, 164:2055–2066. Tan FK, Tercero GM, Arnett FC, et al.: Examination of the possible role of biologically relevant genes around FBN1 in systemic sclerosis in the Choctaw population. Arthritis Rheum 2003, 48:3295–3296. Wallis DD, Tan FK, Kessler R, et al.: Fibrillin 1 abnormalities in dermal fibroblast cultures from first-degree relatives of patients with systemic sclerosis (Scleroderma). Arthritis Rheum 2004, 50:329–332. Yamamoto T, Nishioka K: Possible role of apoptosis in the pathogenesis of bleomycin-induced scleroderma. J Invest Dermatol 2004, 122: 44–50. Yang JW, Mang XH, Li YJ, et al.: Downregulation of Smad transcriptional corepressors SnoN and ski in the fibrotic kidney: An amplification mechanism for TGF-beta 1 signaling. J Am Soc Nephrol 2003, 14:3167+. Zuscik MJ, Rosier RN, Schwarz EM: Altered negative regulation of transforming growth factor beta signaling in scleroderma: Potential involvement of SMURF2 in disease. Arthritis Rheum 2003, 48:1779–1780. tenDijke P, Hill CS: New insights into TGF-betaSmad signalling. Trends Biochem Sci 2004, • 29:265–273. [11]. Animal models: insights into pathogenesis, genetic risk factors, potential therapeutic approaches Review (pp 746–752) Daniel C, Takabatake Y, Mizui M, et al.: Antisense oligonucleotides against thrombospondin-1 inhibit activation of TGF-beta in fibrotic renal disease in the rat in vivo. Am J Pathol 2003, 163:1185–1192. Dekel R, Zvibel I, Brill S, et al.: Gliotoxin ameliorates development of fibrosis and cirrhosis in a thioacetamide rat model. Dig Dis Sci 2003, 48: 1642–1647. HackerFoegen MK, Janson M, Amagai M, et al.: Pathogenicity and epitope characteristics of anti-desmoglein-1 from pemphigus foliaceus patients expressing only IgG1 autoantibodies. J Invest Dermatol 2003, 121:1373–1378. Lemaire R, Farina G, Kissin E, et al.: Mutant fibrillin 1 from tight skin mice increases extracellular matrix incorporation of microfibril-associated glycoprotein 2 and type I collagen. Arthritis Rheum 2004, 50:915–926. Li XP, Rayford H, Uhal BD: Essential roles for angiotensin receptor AT1a in bleomycin-induced apoptosis and lung fibrosis in mice. Am J Pathol 2003, 163:2523–2530. LopezHoyos M, Marquina R, Tamayo E, et al.: Defects in the regulation of B cell apoptosis are required for the production of citrullinated peptide autoantibodies in mice. Arthritis Rheum 2003, 48:2353–2361. Ma LJ, Yang HC, Gaspert A, et al.: Transforming growth factor-b-dependent and -independent pathways of induction of tubulointerstitial fibrosis in beta 6-/- mice. Am J Pathol 2003, 163: 1261–1273. McGaha TL, Le MH, Kodera T, et al.: Molecular mechanisms of interleukin-4 - Induced upregulation of type I collagen gene expression in murine fibroblasts. Arthritis Rheum 2003, 48:2275–2284. Ong VH, Evans LA, Xu SW, et al.: Monocyte chemoattractant protein 3 as a mediator of fibrosis - Overexpression in systemic sclerosis and the type 1 tight-skin mouse. Arthritis Rheum 2003, 48:1979–1991. Ponten A, Li XR, Thoren P, et al.: Transgenic overexpression of platelet-derived growth factor-C in the mouse heart induces cardiac fibrosis, hypertrophy, and dilated cardiomyopathy. Am J Pathol 2003, 163:673–682. Ruzek MC, Jha S, Ledbetter S, et al.: A modified model of graft-versus-host-induced systemic sclerosis (Scleroderma) exhibits all major aspects of the human disease. Arthritis Rheum 2004, 50:1319–1331. Schulz A, Standke D, Kovacevic L, et al.: A major gene locus links early onset albuminuria with renal interstitial fibrosis in the MWF rat with polygenetic albuminuria. J Am Soc Nephrol 2003, 14:3081–3089. Shoshan Y, Mevorach D: Accelerated autoimmune disease in MRL/MpJ-Fas(Lpr) but not in MRL/MpJ following immunization with high load of syngeneic late apoptotic cells. Autoimmunity 2004, 37:103–109. Takagawa S, Lakos G, Mori Y, et al.: Sustained activation of fibroblast transforming growth Factorbeta/Smad signaling in a murine model of scleroderma. J Invest Dermatol 2003, 121: 41–50. Tian B, Han L, Kleidon J, et al.: An HSV-TK transgenic mouse model to evaluate elimination of fibroblasts for fibrosis therapy. Am J Pathol 2003, 163:789–801. Wooley PH: The usefulness and the limitations of animal models in identifying targets for therapy in arthritis. Best Pract Res Clin Rheumatol 2004, 18:47–58. Zhang Y, McCormick LL, Desai SR, et al.: Murine sclerodermatous graft-versus-host disease, a model for human scleroderma - Cutaneous cytokines, chemokines, and immune cell activation. J Immunol 2002, 168:3088–3098. Zhou G, Fujio K, Sadakata A, et al.: Identification of systemically expanded activated T cell clones in MRL/lpr and NZB/W F1 lupus model mice. Clin Exp Immunol 2004, 136:448–455. Recent advances in the pathophysiology, diagnosis, staging, and management of pulmonary hypertension in scleroderma/SSc Allanore Y, Borderie D, Meune C, et al.: N-terminal pro-brain natriuretic peptide as a diagnostic marker of early pulmonary artery hypertension in patients with systemic sclerosis and effects of calcium-channel blockers. Arthritis Rheum 2003, 48:3503–3508. Chang B, Wigley FM, White B, et al.: Scleroderma patients with combined pulmonary hypertension and interstitial lung disease. J Rheumatol 2003, 30:2398–2405. Mukerjee D, StGeorge D, Coleiro B, et al.: Prevalence and outcome in systemic sclerosis associated pulmonary arterial hypertension: application of a registry approach. Ann Rheum Dis 2003, 62:1088–1093. Rosenkranz S, Diet F, Karasch T, et al.: Sildenafil improved pulmonary hypertension and peripheral blood flow in a patient with scleroderma associated lung fibrosis and the Raynaud phenomenon. Ann Intern Med 2003, 139:871– 873. Rosenkranz S, Diet F, Karasch T, et al.: Sildenafil improved pulmonary hypertension and peripheral blood flow in a patient with sclerodermaassociated lung fibrosis and the Raynaud phenomenon. Ann Intern Med 2003, 139:871– 873. Anti-TGF-beta therapy in fibrosis and scleroderma Asano Y, Ihn H, Yamane K, et al.: Impaired Smad7Smurf-mediated negative regulation of TGFbeta signaling in scleroderma fibroblasts. J Clin Invest 2004, 113:253–264. Dai CS, Liu YH: Hepatocyte growth factor antagonizes the profibrotic action of TGF-beta 1 in mesangial cells by stabilizing smad transcriptional corepressor TGIF. J Am Soc Nephrol 2004, 15:1402–1412. ElAgroudy AE, Hassan NA, Foda MA, et al.: Effect of angiotensin II receptor blocker on plasma levels of TGF-beta 1 and interstitial fibrosis in hypertensive kidney transplant patients. Am J Nephrol 2003, 23:300–306. Li Y, Foster W, Deasy BM, et al.: Transforming growth factor-beta 1 induces the differentiation of myogenic cells into fibrotic cells in injured skeletal muscle - A key event in muscle fibrogenesis. Am J Pathol 2004, 164:1007– 1019. Pannu J, Gorehyer E, Yamanaka M, et al.: An increased transforming growth factor beta receptor type I:Type II ratio contributes to elevated collagen protein synthesis that is resistant to inhibition via a kinase-deficient transforming growth factor beta receptor type II in scleroderma. Arthritis Rheum 2004, 50: 1566–1577. Postigo AA: Targeted disruption of TGFbeta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J Clin Invest 2003, 112:1486––1494. Shehata M, Schwarzmeier JD, Hilgarth M, et al.: TGF-beta 1 induces bone marrow reticulin fibrosis in hairy cell leukemia. J Clin Invest 2004, 113:676–685. Shephard P, Martin G, SmolaHess S, et al.: Myofibroblast differentiation is induced in keratinocyte-fibroblast co-cultures and is antagonistically regulated by endogenous transforming growth factor-beta and interleukin-1. Am J Pathol 2004, 164:2055–2066. Treatment Allanore Y, Borderie D, Lemarechal H, et al.: Acute and sustained effects of dihydropyridine-type calcium channel antagonists on oxidative stress in systemic sclerosis. Am J Med 2004, 116:595–600. Kimura T, Ishii Y, Morishima Y, et al.: Treatment with alpha-galactosylceramide attenuates the development of bleomycin-induced pulmonary fibrosis. J Immunol 2004, 172:5782–5789. Levy Y, Amital H, Langevitz P, et al.: Intravenous immunoglobulin modulates cutaneous involvement and reduces skin ribrosis in systemic sclerosis: an open-label study. Arthritis Rheum 2004, 50:1005–1007. Mayes MD, ODonnell D, Rothfield NF, et al.: Minocycline is not effective in systemic sclerosis Results of an open-label multicenter trial. Arthritis Rheum 2004, 50:553–557. Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Renal fibrosis 771 Shoenfeld Y, Krause I: IVIG for autoimmune, fibrosis, and malignant conditions: Our experience with 200 patients. J Clin Immunol 2004, 24: 107–114. Renal fibrosis Eis V, Luckow B, Vielhauer V, et al.: Chemokine receptor CCR1 but not CCR5 mediates leukocyte recruitment and subsequent renal fibrosis after unilateral ureteral obstruction. J Am Soc Nephrol 2004, 15:337–347. Gross O, Beirowski B, Keopke ML, et al.: Preemptive ramipril therapy delays renal failure and reduces renal fibrosis in COL4A3-knockout mice with A1port syndrome. Kidney Int 2003, 63:438–446. Maric C, Sandberg K, HinojosaLaborde C: Glomerulosclerosis and tubulointerstitial fibrosis are attenuated with 17b-estradiol in the aging Dahl salt sensitive rat. J Am Soc Nephrol 2004, 15:1546–1556. Masterson R, Hewitson TD, Kelynack K, et al.: Relaxin down-regulates renal fibroblast function and promotes matrix remodelling in vitro. Nephrol Dial Transplant 2004, 19:544–552. Postigo AA: Targeted disruption of TGFbeta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J Clin Invest 2003, 112:1486––1494. Rouschop KMA, Sewnath ME, Claessen N, et al.: CD44 deficiency increases tubular damage but reduces renal fibrosis in obstructive nephropathy. J Am Soc Nephrol 2004, 15:674– 686. Stambe C, Atkins RC, Tesch GH, et al.: The role of p38 alpha mitogen-activated protein kinase activation in renal fibrosis. J Am Soc Nephrol 2004, 15:370–379. Wada T, Furuichi K, Sakai N, et al.: Gene therapy via blockade of monocyte chemoattractant protein-1 for renal fibrosis. J Am Soc Nephrol 2004, 15:940–948. Hepatic fibrosis Hattori N, Mizuno S, Yoshida Y, et al.: The plasminogen activation system reduces fibrosis in the lung by a hepatocyte growth factor-dependent mechanism. Am J Pathol 2004, 164:1091– 1098. Zeitoun D, Brancatelli G, Colombat M, et al.: Congenital hepatic fibrosis: CT findings in 18 adults. Radiology 2004, 231:109–116. SLE AlMaini MH, ElAgeb EM, AlWahaibi SS, et al.: Demographic, autoimmune, and clinical profiles of patients with systemic lupus erythematosus in Oman. Rheumatol Int 2003, 23:186–191. Alves JD, Kumar S, Isenberg DA: Cross-reactivity between anti-cardiolipin, anti-high-density lipoprotein and anti-apolipoprotein A-I IgG antibodies in patients with systemic lupus erythematosus and primary antiphospholipid syndrome. Rheumatology 2003, 42:893–899. Bujan S, OrdiRos J, Paredes J, et al.: Contribution of the initial features of systemic lupus erythematosus to the clinical evolution and survival of a cohort of Mediterranean patients. Ann Rheum Dis 2003, 62:859–865. Chauhan R, Handa R, Das TP, et al.: Over-expression of TATA binding protein (TBP) and p53 and autoantibodies to these antigens are features of systemic sclerosis, systemic lupus erythematosus and overlap syndromes. Clin Exp Immunol 2004, 136:574–584. Constans J, Dupuy R, Blann AD, et al.: Anti-endothelial cell autoantibodies and soluble markers of endothelial cell dysfunction in systemic lu- pus erythematosus. J Rheumatol 2003, 30: 1963–1966. Cuchacovich R, Espinoza LR: Clinical and serological associations of ribosomal P autoantibodies in systemic lupus erythematosus: prospective evaluation in a large cohort of Italian patients. Rheumatology 2003, 42:1115–1116. Feist E, Keitzer R, Gerhold K, et al.: Development of systemic lupus erythematosus in a patient with congenital heart block. Arthritis Rheum 2003, 48:2697–2698. GomezZumaquero JM, Tinahones FJ, DeRamon E, et al.: Association of biological markers of activity of systemic lupus erythematosus with levels of anti-oxidized low-density lipoprotein antibodies. Rheumatology 2004, 43:510–513. Hamasaki K, Mimura T, Kanda H, et al.: Systemic lupus erythematosus and thrombotic thrombocytopenic purpura: a case report and literature review. Clin Rheumatol 2003, 22:355–358. Ho HH, Lin JL, Wu YJJ, et al.: Gout in systemic lupus erythematosus and overlap syndrome - a hospital-based study. Clin Rheumatol 2003, 22:295–298. Ignat GP, Rat AC, Sychra JJ, et al.: Information on diagnosis and management of systemic lupus erythematosus derived from the routine measurement of 8 nuclear autoantibodies. J Rheumatol 2003, 30:1761–1769. Mannik M, Merrill CE, Stamps LD, et al.: Multiple autoantibodies form the glomerular immune deposits in patients with systemic lupus erythematosus. J Rheumatol 2003, 30:1495– 1504. Mason LJ, Ravirajan CT, Rahman A, et al.: Is alphaactinin a target for pathogenic anti-DNA antibodies in lupus nephritis? Arthritis Rheum 2004, 50:866–870. Noel B: Statins and lupus erythematosus. Rheumatology 2004, 43:397–398. Oelzner P, Deliyska B, Funfstuck R, et al.: Anti-C1q antibodies and antiendothelial cell antibodies in systemic lupus erythematosus - relationship with disease activity and renal involvement. Clin Rheumatol 2003, 22:271–278. Otsuka J, Horiuchi T, Yoshizawa S, et al.: Association of a four-amino acid residue insertion polymorphism of the HS1 gene with systemic lupus erythematosus - Molecular and functional analysis. Arthritis Rheum 2004, 50:871–881. Perlemuter G, Cacoub P, Sbai A, et al.: Hepatitis C virus infection in systemic lupus erythematosus: A case-control study. J Rheumatol 2003, 30:1473–1478. Seelen MA, Trouw LA, VanderHoorn JWA, et al.: Autoantibodies against mannose-binding lectin in systemic lupus erythematosus. Clin Exp Immunol 2003, 134:335–343. Simon JA, Cabiedes J, Ortiz E, et al.: Anti-nucleosome antibodies in patients with systemic lupus erythematosus of recent onset. Potential utility as a diagnostic tool and disease activity marker. Rheumatology 2004, 43:220–224. Sjowall C, Bengtsson AA, Sturfelt G, et al.: Serum levels of autoantibodies against monomeric C-reactive protein are correlated with disease activity in systemic lupus erythematosus. Arthritis Res Ther 2004, 6:R87–R94. Takahashi R, Tsutsumi A, Ohtani K, et al.: Anti-mannose binding lectin antibodies in sera of Japanese patients with systemic lupus erythematosus. Clin Exp Immunol 2004, 136:585–590. VeraLastra O, Mendez C, Jara LJ, et al.: Correlation of prolactin serum concentrations with clinical activity and remission in patients with systemic lupus erythematosus. Effect of conventional treatment. J Rheumatol 2003, 30:2140–2146. Wajed J, Ahmad Y, Durrington PN, et al.: Statins and lupus erythematosus - Reply. Rheumatology 2004, 43:398–399. Miscellaneous Adami S, Braga V, Zamboni M, et al.: Relationship between lipids and bone mass in 2 cohorts of healthy women and men. Calcif Tissue Int 2004, 74:136–142. Adams KM, Lambert NC, Heimfeld S, et al.: Male DNA in female donor apheresis and CD34enriched products. Blood 2003, 102:3845– 3847. Akaogi J, Nacionales DC, Kuroda Y, et al.: Ecotropic murine leukemia viruses and exogenous mouse mammary tumor viruses are not essential for pristane-induced lupus. Arthritis Rheum 2003, 48:2990–2992. Akdis CA, Kussebi F, Pulendran B, et al.: Inhibition of T helper 2-type responses, IgE production and eosinophilia by synthetic lipopeptides. Eur J Immunol 2003, 33:2717–2726. Akesson A, Fiori G, Krieg T, et al.: Assessment of skin, joint, tendon and muscle involvement. Clin Exp Rheumatol 2003, 21:S5–S8. Allcock RJ, Forrest I, Corris PA, et al.: A study of the prevalence of systemic sclerosis in northeast England. Rheumatology 2004, 43:596– 602. Anderson JL, Muhlestein JB: Restenosis after coronary intervention: Narrowing C-reactive protein’s prognostic potential? Am J Med 2003, 115:147–149. Appenzeller S, Montenegro MA, Dertkigil SS, et al.: Neuroimaging findings in scleroderma en coup de sabre. Neurology 2004, 62:1585–1589. Apras S, Ertenli I, Ozbalkan Z, et al.: Effects of oral cyclophosphamide and prednisolone therapy on the endothelial functions and clinical findings in patients with early diffuse systemic sclerosis. Arthritis Rheum 2003, 48:2256– 2261. Ardic F, Soyupek F, Kahraman Y, et al.: The musculoskeletal complications seen in type II diabetics: predominance of hand involvement. Clin Rheumatol 2003, 22:229–233. Arenas JI, GallegosOrozco JF, Laskus T, et al.: Hepatitis C virus quasi-species dynamics predict progression of fibrosis after liver transplantation. J Infect Dis 2004, 189:2037–2046. Arif A, Levine RA, Sanderson SO, et al.: Regression of fibrosis in chronic hepatitis C after therapy with interferon and ribavirin. Dig Dis Sci 2003, 48:1425–1430. Arranz I, MartinSuarez A, Lanao JM, et al.: Population pharmacokinetics of high dose ibuprofen in cystic fibrosis. Arch Dis Child 2003, 88: 1128–1130. Asherson RA, Shoenfeld Y, Bosman C: AD unusually complicated case of primary Sjogren’s syndrome: development of transient “lupustype” autoantibodies following silicone implant rejection (Vol 31, pg 196, 2004). J Rheumatol 2004, 31:405. Asherson RA, Shoenfeld Y: Annular, semicircular lipoatrophy of the thighs associated with incomplete ”CREST” syndrome - Raynaud’s, telangiectasias, and esophageal dysmotility. JCR - J Clin Rheumatol 2003, 9:129–133. Atamas SP: FCP (Http://fibro.biobitfield.com/fcp.php): a bioinformatic tool assisting in PubMed searches for literature on fibrosis-related cytokines. Arthritis Rheum 2003, 48:2083–2084. Aziz KA, Till KJ, Chen HJ, et al.: The role of autocrine FGF-2 in the distinctive bone marrow fibrosis of hairy-cell leukemia (HCL). Blood 2003, 102:1051–1056. Aznarez I, Chan EM, Zielenski J, et al.: Characterization of disease-associated mutations affecting an exonic splicing enhancer and two cryptic splice sites in exon 13 of the cystic fibrosis transmembrane conductance regulator gene. Hum Mol Genet 2003, 12:2031–2040. Bajaj J, Knox JF, Komorowski R, et al.: The irony of herbal hepatitis - Ma-Huang-induced hepatotoxicity associated with compound heterozygosity for hereditary hemochromatosis. Dig Dis Sci 2003, 48:1925–1928. BalfourLynn IM, Mohan U, Bush A, et al.: Intravenous immunoglobulin for cystic fibrosis lung 772 Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Miscellaneous disease: a case series of 16 children. Arch Dis Child 2004, 89:315–319. Ballantyne CM, Davidson MH: Possible differences between fibrates in pharmacokinetic interactions with statins. Arch Intern Med 2003, 163: 2394–2395. Ballantyne CM, Corsini A, Davidson MH, et al.: Risk for myopathy with statin therapy in high risk patients. Arch Intern Med 2003, 163:553– 564. BarMeir E, Eherenfeld M, Shoenfeld Y: Silicone gel breast implants and connective tissue disease - A comprehensive review. Autoimmunity 2003, 36:193–197. Bataller R, Schwabe RF, Choi YH, et al.: NADPH oxidase signal transduces angiotensin II in hepatic stellate cells and is critical in hepatic fibrosis. J Clin Invest 2003, 112:1383–1394. Bauer DC, Mundy GR, Jamal SA, et al.: Use of statins and fracture - Results of 4 prospective studies and cumulative meta-analysis of observational studies and controlled trials. Arch Intern Med 2004, 164:146–152. Baumer JH: Evidence based guidelines for the performance of the sweat test for the investigation of cystic fibrosis in the UK. Arch Dis Child 2003, 88:1126–1127. Bazzichi L, Rossi A, DeGiorgio F, et al.: Clinical improvement in systemic sclerosis resulting from urokinase therapy explained by light and electron microscopy skin examination. Scand J Rheumatol 2003, 32:261–267. Bernacchi E, Amato L, Parodi A, et al.: Sjogren’s syndrome: A retrospective review of the cutaneous features of 93 patients by the Italian Group of Immunodermatology. Clin Exp Rheumatol 2004, 22:55–62. Bertinotti L, Bracci S, Nacci F, et al.: The autonomic nervous system in systemic sclerosis. A review. Clin Rheumatol 2004, 23:1–5. Blank M, Krause I, Fridkin M, et al.: Baterial induction of autoantibodies to beta2-glycoprotein-I accounts for the infectious etiology of antiphospholipid syndrome. J Clin Invest 2002, 109:797–804. Blaszczyk M, Krolicki L, Krasu M, et al.: Progressive facial hemiatrophy: Central nervous system involvement and relationship with scleroderma en coup de sabre. J Rheumatol 2003, 30:1997–2004. Boackle SA, Culhane KK, Brown JM, et al.: CR1/CR2 deficiency alters IgG3 autoantibody production and IgA glomerular deposition in the MRL/lpr model of SLE. Autoimmunity 2004, 37:111–123. Bohm I: Flow cytometric analysis of the LE cell phenomenon. Autoimmunity 2004, 37:37–44. Bombardieri S, Medsger TA, Silman AJ, et al.: The assessment of the patient with systemic sclerosis - Introduction. Clin Exp Rheumatol 2003, 21:S2–S4. Boon RL, Baildam E: Progressive fibrosis of the quadriceps muscle. Arch Dis Child 2004, 89:44. Bourquin C, Schubart A, Tobollik S, et al.: Selective unresponsiveness to conformational B cell epitopes of the myelin oligodendrocyte glycoprotein in H-2(B) mice. J Immunol 2003, 171: 455–461. Boyman O, Hefti HP, Conrad C, et al.: Spontaneous development of psoriasis in a new animal model shows an essential role for resident T cells and tumor necrosis factor-alpha. J Exp Med 2004, 199:731–736. Brittenham GM, Nathan DG, Olivieri NF, et al.: Deferiprone and hepatic fibrosis. Blood 2003, 101:5089–5090. Brody LC, Conley M, Cox C, et al.: A polymorphism, R653Q, in the trifunctional enzyme methylenetetrahydrofolate dehydrogenase/methenyltetrahydrofolate cyclohydrolase/formyltetrahydrofolate synthetase is a maternal genetic risk factor for neural tube defects - Report of the Birth Defects Re- search Group. Am J Hum Genet 2002, 71: 1207–1215. Brokken LJS, Wiersinga WM, Prummel MF: Thyrotropin receptor autoantibodies are associated with continued thyrotropin suppression in treated euthyroid Graves’ disease patients. J Clin Endocrinol Metab 2003, 88:4135–4138. Buchman AL, Rao SM: A patient with a polyglandular autoimmune syndrome involving the salivary glands, thyroid, intestine, and pancreas. Dig Dis Sci 2004, 49:590–593. Burch M, Aurora P: Current status of paediatric heart, lung, and heart-lung transplantation. Arch Dis Child 2004, 89:386–389. Busser BW, Adalr BS, Erikson J, et al.: Activation of diverse repertoires of autoreactive T cells enhances the loss of anti-dsDNA B cell tolerance. J Clin Invest 2003, 112:1361–1371. Button BM, Heine RG, CattoSmith AG, et al.: Chest physiotherapy, gastro-oesophageal reflux, and arousal in infants with cystic fibrosis. Arch Dis Child 2004, 89:435–439. Byrne NM, Keavey PM, Perry JD, et al.: Comparison of lung deposition of colomycin using the HaloLite and the Pari LC Plus nebulisers in patients with cystic fibrosis. Arch Dis Child 2003, 88:715–718. Calguneri M, Apras S, Ozbalkan Z, et al.: The efficacy of oral cyclophosphamide plus prednisolone in early diffuse systemic sclerosis. Clin Rheumatol 2003, 22:289–294. Calo LA, Zaghetto F, Pagnin E, et al.: Effect of aldosterone and glycyrrhetinic acid on the protein expression of PAI-1 and p22(Phox) in human mononuclear leukocytes. J Clin Endocrinol Metab 2004, 89:1973–1976. Canbay A, Guicciardi ME, Higuchi H, et al.: Cathepsin B inactivation attenuates hepatic injury and fibrosis during cholestasis. J Clin Invest 2003, 112:152–159. Cannick L, Douglas G, Crater S, et al.: Nodular scleroderma: Case report and literature review. J Rheumatol 2003, 30:2500–2502. Canto E, Vidal S, RodriguezSanchez JL: HKATPase expression in the susceptible BALB/c and the resistant DBA/2 strains of mice to autoimmune gastritis. Autoimmunity 2003, 36: 275–283. Carroccio A, Volta U, DiPrima L, et al.: Autoimmune enteropathy and colitis in an adult patient. Dig Dis Sci 2003, 48:1600–1606. Carroccio A, Iannitto E, DiPrima L, et al.: Screening for Celiac disease in non-Hodgkin’s lymphoma patients - A serum anti-transglutaminasebased approach. Dig Dis Sci 2003, 48:1530– 1536. Caserta L, deMagistris L, Secondulfo M, et al.: Assessment of intestinal permeability and orocecal transit time in patients with systemic sclerosis: analysis of relationships with epidemiologic and clinical parameters. Rheumatol Int 2003, 23:226–230. Cavill D, Waterman SA, Gordon TP: Antiidiotypic antibodies neutralize autoantibodies that inhibit cholinergic neurotransmission. Arthritis Rheum 2003, 48:3597–3602. Chambler AFW, Carr AJ: The role of surgery in frozen shoulder. J Bone Joint Surg (Br) 2003, 85B:789–795. Chan ATY, Wordsworth BP, McNally J: Overlap connective tissue disease, pulmonary fibrosis, and extensive subcutaneous calcification. Ann Rheum Dis 2003, 62:690–691. Chang M, Nakagawa PA, Williams SA, et al.: Immune thrombocytopenic purpura (ITP) plasma and purified ITP monoclonal autoantibodies inhibit megakaryocytopoiesis in vitro. Blood 2003, 102:887–895. Charytan DM, Le DD, Grossman JM, et al.: Antiglomerular basement membrane autoantibodies are nonpathogenic in Wegener’s granulomatosis. Am J Med 2003, 115:414–415. Chazenbalk GD, Pichurin P, Chen CR: Thyroid stimulating autoantibodies in Graves’ disease preferentially recognize the free A subunit, not the thyrotropin holoreceptor. J Clin Invest 2002, 110:209–217. Chazenbalk GD, Latrofa F, McLachlan SM, et al.: Thyroid stimulation does not require antibodies with identical epitopes but does involve recognition of a critical conformation at the N terminus of the thyrotropin receptor A-subunit. J Clin Endocrinol Metab 2004, 89:1788–1793. Chen CR, Pichurin P, Nagayama Y, et al.: The thyrotropin receptor autoantigen in Graves disease is the culprit as well as the victim. J Clin Invest 2003, 111:1897–1904. Chevillard C, Moukoko CE, Elwali NEMA, et al.: IFN-gamma polymorphisms (IFNgamma+2109 and IFN-gamma+3810) are associated with severe hepatic fibrosis in human hepatic schistosomiasis (Schistosoma mansoni). J Immunol 2003, 171:5596–5601. Choi JJ, Min DJ, Cho ML, et al.: Elevated vascular endothelial growth factor in systemic sclerosis. J Rheumatol 2003, 30:1529–1533. ChristopherStine L, Wigley F: Tumor necrosis factor-alpha antagonists induce lupus-like syndrome in patients with scleroderma overlap/mixed connective tissue disease. J Rheumatol 2003, 30:2725–2727. Church TS, Earnest CP, Wood KA, et al.: Reduction of C-reactive protein levels through use of a multivitamin. Am J Med 2003, 115:702– 707. Chvatchko Y, Proudfoot AEI, Buser R, et al.: Inhibition of airway inflammation by amino-terminally modified RANTES/CC chemokine ligand 5 analogues is not mediated through CCR3. J Immunol 2003, 171:5498–5506. Citores MJ, RuaFigueroa I, RodriguezGallego C, et al.: The dinucleotide repeat polymorphism in the 3’UTR of the CD154 gene has a functional role on protein expression and is associated with systemic lupus erythematosus. Ann Rheum Dis 2004, 63:310–317. Citron N, Hearnden A: Skin tension in the aetiology of Dupuytren’s disease; A prospective trial. J Hand Surg [Br and Eur] 2003, 28B:528–530. Clancy RM, Chan EKL, Buyon JP: Does the cellular localization of antigens in or on apoptotic blebs influence the pathogenicity or benefit of cognate antibodies? Comment on the article by Dieude et al. Arthritis Rheum 2003, 48: 2080–2082. Clark H, Reid K: The potential of recombinant surfactant protein D therapy to reduce inflammation in neonatal chronic lung disease, cystic fibrosis, and emphysema. Arch Dis Child 2003, 88:981–984. Cohen JS: Statins and low-density lipoprotein cholesterol levels. Am J Med 2003, 115:74–75. Colombo M, Dono M, Gazzola P, et al.: Maintenance of B lymphocyte-related clones in the cerebrospinal fluid of multiple sclerosis patients. Eur J Immunol 2003, 33:3433–3438. Connolly SB, Sadlier D, Kieran NE, et al.: Transcriptome profiling and the pathogenesis of diabetic complications. J Am Soc Nephrol 2003, 14:S279–S283. Corbin IR, Minuk GY: Serial percutaneous liver biopsies in laboratory rats. Dig Dis Sci 2003, 48:1939–1943. Cugno M, Cabibbe M, Galli M, et al.: Antibodies to tissue-type plasminogen activator (TPA) in patients with antiphospholipid syndrome: evidence of interaction between the antibodies and the catalytic domain of tPA in 2 patients. Blood 2004, 103:2121–2126. DAngelo S, Cuomo G, Paone C, et al.: Low-dose intravenous cyclophosphamide in systemic sclerosis: a preliminary safety study. Clin Rheumatol 2003, 22:393–396. Dandona P, Kumar V, Aljada A, et al.: Angiotensin II receptor blocker valsartan suppresses reactive oxygen species generation in leukocytes, nuclear factor-kappa B, in mononuclear cells of normal subjects: Evidence of an antiinflammatory action. J Clin Endocrinol Metab 2003, 88:4496–4501. Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Miscellaneous 773 Daniel V, Sadeghi M, Naujokat C, et al.: Evidence for autoantibody-induced CD4 depletion mediated by apoptotic and non-apoptotic mechanisms in HIV-positive long-term surviving haemophilia patients. Clin Exp Immunol 2004, 135:94–104. Daniels JT, Schultz GS, Blalock TD, et al.: Mediation of transforming growth factor-beta(1)stimulated matrix contraction by fibroblasts - A role for connective tissue growth factor in contractile scarring. Am J Pathol 2003, 163:2043–2052. Daskalopoulou SS, Mikhailidis DP, Athyros VG, et al.: Fenofibrate and losartan. Ann Rheum Dis 2004, 63:469–470. DelPuente A, Esposito A, Savastano S, et al.: Rheumatoid arthritis and thyroid disease - Reply. Clin Exp Rheumatol 2004, 22:265–266. DelRosso A, Bertinotti L, Pietrini U, et al.: Pupillocynetic activity of substance P in systemic sclerosis. J Rheumatol 2003, 30:1231–1237. Dewald O, Ren GF, Duerr GD, et al.: Of mice and dogs: Species-specific differences in the inflammatory response following myocardial infarction. Am J Pathol 2004, 164:665–677. Dibra A, Mehilli J, Braun S, et al.: Association between C-reactive protein levels and subsequent cardiac events among patients with stable angina treated with coronary artery stenting. Am J Med 2003, 114:715–722. Dixit K, Ahsan H, Ali A: Polydeoxyribonucleotide C photoconjugated with lysine or arginine present unique epitopes for human anti-DNA autoantibodies. Hum Immunol 2003, 64:880– 886. Dunsmore SE, Shapiro SD: The bone marrow leaves its scar: new concepts in pulmonary fibrosis. J Clin Invest 2004, 113:180–182. Dziadzio M, Giovagnoni A, Pomponio G, et al.: Acute myocarditis associated with adenoviral infection in a patient with scleroderma. Clin Rheumatol 2003, 22:487–490. Dziadzio M, Smith RE, Abraham DJ, et al.: Serological assessment of type I collagen burden in scleroderma spectrum disorders: A systematic review. Clin Exp Rheumatol 2004, 22:356– 367. Egawa N, Irie T, Tu YY, et al.: A case of autoimmune pancreatitis with initially negative autoantibodies turning positive during the clinical course. Dig Dis Sci 2003, 48:1705–1708. Egle A, Harris AW, Bath ML, et al.: VavP-Bcl2 transgenic mice develop follicular lymphoma preceded by germinal center hyperplasia. Blood 2004, 103:2276–2283. Eisenbarth SC, Piggott DA, Bottomly K: The master regulators of allergic inflammation: dendritic cells in Th2 sensitization. Curr Opin Immunol 2003, 15:620–626. Elfving AM, Lindberg BA, Nystrom L, et al.: Islet autoantibodies in cord blood from patients who developed type 1 diabetes mellitus at 15-30 years of age. Autoimmunity 2003, 36: 227–231. Evenepoel P, Zeegers M, Segaert S, et al.: Nephrogenic fibrosing dermopathy: a novel, disabling disorder in patients with renal failure. Nephrol Dial Transplant 2004, 19:469–473. Evoli A, Tonali PA, Padua L, et al.: Clinical correlates with anti-MuSK antibodies in generalized seronegative myasthenia gravis. Brain 2003, 126:2304–2311. Fagiolo E, TorianiTerenzi C: Mechanisms of immunological tolerance loss versus erythrocyte self-antigens and autoimmune hemolytic anemia. Autoimmunity 2003, 36:199–204. Fan GC, Singh RR: Vaccination with minigenes encoding V (H)-derived major histocompatibility complex class I-binding epitopes activates cytotoxic T cells that ablate autoantibody-producing B cells and inhibit lupus. J Exp Med 2002, 196:731–741. Farre C, DomingoDomenech E, Font R, et al.: Celiac disease and lymphoma risk: A multicentric case-control study in Spain. Dig Dis Sci 2004, 49:408–412. Fenoglio LM, Scalabrino E, Perin PC: Eosinophilic gastroenteritis: Review of the literature - Response. Dig Dis Sci 2003, 48:2215. Ferraccioli GF, Gremese E: Autoantibodies and thrombophilia in RA: TNF alpha and TNF alpha blockers. Ann Rheum Dis 2004, 63:613–615. Ferrara DE, Liu XW, Espinola RG, et al.: Inhibition of the thrombogenic and inflammatory properties of antiphospholipid antibodies by fluvastatin in an in vivo animal model. Arthritis Rheum 2003, 48:3272–3279. Fields ML, Erikson J: The regulation of lupus-associated autoantibodies: immunoglobulin transgenic models. Curr Opin Immunol 2003, 15: 709–717. Fischetti F, Carretta R, Borotto G, et al.: Fluvastatin treatment inhibits leucocyte adhesion and extravasation in models of complement-mediated acute inflammation. Clin Exp Immunol 2004, 135:186–193. FloresAlvarado DE, EsquivelValerio JA, GarzaElizondo M: Linear scleroderma en Coup de Sabre and brain calcification: Is there a pathogenic relationship? Reply. J Rheumatol 2003, 30:2725. Fogo AB: Renal fibrosis: not just PAI-1 in the sky. J Clin Invest 2003, 112:326–328. Foucher G, Medina J, Navarro R: Percutaneous needle aponeurotomy: Complications and results. J Hand Surg [Br and Eur] 2003, 28B: 427–431. FranckLarsson K, Hedenstrom H, Dahl R, et al.: Delayed gastric emptying in patients with diffuse versus limited systemic sclerosis, unrelated to gastrointestinal symptoms and myoelectric gastric activity. Scand J Rheumatol 2003, 32:351–358. Frediani B, Baldi F, Falsetti P, et al.: Bone mineral density in patients with systemic sclerosis. Ann Rheum Dis 2004, 63:326–327. Fredlani B, Baldi F, Falsetti P, et al.: Clinical determinants of bone mass and bone ultrasonometry in patients with systemic sclerosis. Clin Exp Rheumatol 2004, 22:313–318. Frolkis JP, Pearce GL, Sprecher DL: Statins and low-density lipoprotein cholesterol levels - Reply. Am J Med 2003, 115:75–76. Fuchtenbusch M, Bonifacio E, Lampasona V, et al.: Immune responses to glutamic acid decarboxylase and insulin in patients with gestational diabetes. Clin Exp Immunol 2004, 135:318– 321. Furuse S, Fujii H, Kaburagi Y, et al.: Serum concentrations of the CXC chemokines interleukin 8 and growth-regulated oncogene-alpha are elevated in patients with systemic sclerosis. J Rheumatol 2003, 30:1524–1528. Fux R, Morike K, Gundel UF, et al.: Ezetimibe and statin-associated myopathy. Ann Intern Med 2004, 140:671–672. Galle P, Hougs L, Barington T, et al.: Knocking out IL-6 by vaccination. Eur J Immunol 2004, 34: 291–300. Gasparotti R, Zanetti D, Bolzoni A, et al.: Inflammatory myofibroblastic tumor of the temporal bone. AJNR Am J Neuroradiol 2003, 24:2092–2096. Gentile F, Conte M, Formisano S: Thyroglobulin as an autoantigen: what can we learn about immunopathogenicity from the correlation of antigenic properties with protein structure? Immunology 2004, 112:13–25. Ghirardello A, Doria A, Zampieri S, et al.: Antinucleosome antibodies in SLE: a two-year follow-up study of 101 patients. J Autoimmun 2004, 22:235–240. Giles IP, Isenberg DA, Latchman DS, et al.: How do antiphospholipid antibodies bind beta(2)-glycoprotein I? Arthritis Rheum 2003, 48:2111– 2121. Golomb BA, Criqui MH, White H, et al.: Conceptual foundations of the UCSD statin study - A randomized controlled trial assessing the impact of statins on cognition, behavior, and biochemistry. Arch Intern Med 2004, 164:153–162. GomezPuerta JA, Gil V, Cervera R, et al.: Coeliac disease associated with systemic sclerosis. Ann Rheum Dis 2004, 63:104–105. Gora M, Gardas A, Wiktorowicz W, et al.: Evaluation of conformational epitopes on thyroid peroxidase by antipeptide antibody binding and mutagenesis. Clin Exp Immunol 2004, 136: 137–144. Gottenberg JE, Busson M, Loiseau P, et al.: In primary Sjogren’s syndrome, HLA class II is associated exclusively with autoantibody production and spreading of the autoimmune response. Arthritis Rheum 2003, 48:2240– 2245. Gris JC, Perneger TV, Quere I, et al.: Antiphospholipid/antiprotein antibodies, hemostasis-related autoantibodies, and plasma homocysteine as risk factors for a first early pregnancy loss: a matched case-control study. Blood 2003, 102:3504–3513. Groman JD, Hefferon TW, Casals T, et al.: Variation in a repeat sequence determines whether a common variant of the cystic fibrosis transmembrane conductance regulator gene is pathogenic or benign. Am J Hum Genet 2004, 74:176–179. Guis S, Mattei JP, Liote F: Drug-induced and toxic myopathies. Best Pract Res Clin Rheumatol 2003, 17:877–907. GuzylackPiriou L, Balmelli C, McCullough KC, et al.: Type-A CpG oligonucleotides activate exclusively porcine natural interferon-producing cells to secrete interferon-alpha, tumour necrosis factor-alpha and interleukin-12. Immunology 2004, 112:28–37. Gylling M, Kaariainen E, Vaisanen R, et al.: The hypoparathyroidism of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy protective effect of male sex. J Clin Endocrinol Metab 2003, 88:4602–4608. Hakkinen L, Koivisto L, Gardner H, et al.: Increased expression of beta 6-integrin in skin leads to spontaneous development of chronic wounds. Am J Pathol 2004, 164:229–242. Hannallah D, Peng HR, Young B, et al.: Retroviral delivery of Noggin inhibits the formation of heterotopic ossification induced by BMP-(4), Demineralized bone matrix, and trauma in an animal model. J Bone Joint Surg (Am) 2004, 86A: 80–91. Hart DA, Kydd AS, Frank CB, et al.: Tissue repair in rheumatoid arthritis: challenges and opportunities in the face of a systemic inflammatory disease. Best Pract Res Clin Rheumatol 2004, 18:187–202. Hashimoto N, Jin H, Liu TJ, et al.: Bone marrowderived progenitor cells in pulmonary fibrosis. J Clin Invest 2004, 113:243–252. He L, Hu CS, Cai GB, et al.: Highly up-regulated CXCR3 expression on eosinophils in mice infected with Schistosoma japonicum. Immunology 2004, 111:107–117. Hedman MH, Rolandsson O, Hagg E, et al.: Association between insulin resistance and GAD65-autoantibody levels - A pilot study in an adult non-diabetic population. Autoimmunity 2004, 37:33–36. Helmby H, Grencis RK: Contrasting roles for IL-10 in protective immunity to different life cycle stages of intestinal nematode parasites. Eur J Immunol 2003, 33:2382–2390. HernandezPando R, Aguilar D, Hernandez MLG, et al.: Pulmonary tuberculosis in BALB/c mice with nonfunctional IL-4 genes: changes in the inflammatory effects of TNF-alpha and in the regulation of fibrosis. Eur J Immunol 2004, 34: 174–183. Herschman HR: Micro-PET imaging and small animal models of disease. Curr Opin Immunol 2003, 15:378–384. Hill CL: Increased risk of cancer in patients with scleroderma: no risk in patients with morphoea? Reply. Ann Rheum Dis 2004, 63:612. 774 Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Miscellaneous Hill CL, Nguyen AM, Roder D, et al.: Risk of cancer in patients with scleroderma: a population based cohort study. Ann Rheum Dis 2003, 62:728–731. Hirano N, Butler MO, vonBergweltBaildon MS, et al.: Autoantibodies frequently detected in patients with aplastic anemia. Blood 2003, 102: 4567–4575. Hirsh M, Dyugovskaya L, Kaplan V, et al.: Response of lung gamma delta T cells to experimental sepsis in mice. Immunology 2004, 112:153– 160. Hocevar A, Tomsic M, Praprotnik S, et al.: Parasympathetic nervous system dysfunction in primary Sjogren’s syndrome. Ann Rheum Dis 2003, 62:702–704. Hoflich C, Sabat R, Rosseau S, et al.: Naturally occurring anti-IFN-gamma autoantibody and severe infections with Mycobacterium cheloneae and Burkholderia cocovenenans. Blood 2004, 103:673–675. Holdcraft LC, Assefi N, Buchwald D: Complementary and alternative medicine in fibromyalgia and related syndromes. Best Pract Res Clin Rheumatol 2003, 17:667–683. Holsti MA, Chitnis T, Panzo RJ, et al.: Regulation of postsurgical fibrosis by the programmed death-1 inhibitory pathway. J Immunol 2004, 172:5774–5781. Hong P, Pope JE, Ouimet JM, et al.: Erectile dysfunction associated with scleroderma: A casecontrol study of men with scleroderma and rheumatoid arthritis. J Rheumatol 2004, 31: 508–513. Hoppu S, Ronkainen MS, Kulmala P, et al.: GAD65 antibody isotypes and epitope recognition during the prediabetic process in siblings of children with type I diabetes. Clin Exp Immunol 2004, 136:120–128. Huang SY, Tien HF, Su FH, et al.: Nonirradiated NOD/SCID-human chimeric animal model for primary human multiple myeloma - A potential in vivo culture system. Am J Pathol 2004, 164: 747–756. Huaux F, Liu TJ, McGarry B, et al.: Eosinophils and T lymphocytes possess distinct roles in bleomycin-induced lung injury and fibrosis. J Immunol 2003, 171:5470–5481. Hubeau C, LeNaour R, Abely M, et al.: Dysregulation of IL-2 and IL-8 production in circulating T lymphocytes from young cystic fibrosis patients (Vol 135, pg 528, 2004). Clin Exp Immunol 2004, 136:188. Hueber W, Utz PJ, Robinson WH: Autoantibodies in early arthritis: Advances in diagnosis and prognostication. Clin Exp Rheumatol 2003, 21:S59–S64. Hufnagl K, Wagner B, Winkler B, et al.: Induction of mucosal tolerance with recombinant Hev b 1 and recombinant Hev b 3 for prevention of latex allergy in BALB/c mice. Clin Exp Immunol 2003, 133:170–176. Hummel M, Bonifacio E, Schmid S, et al.: Brief communication: Early appearance of islet autoantibodies predicts childhood type 1 diabetes in offspring of diabetic parents. Ann Intern Med 2004, 140:882–886. Hussain SZ, Chu C, Greenberg DP, et al.: Clostridium difficile colitis in children with cystic fibrosis. Dig Dis Sci 2004, 49:116–121. Ivanovska N, Yordanov M, Raykovska V: Single immunization of newborn mice with heterologous type-II collagen induces arthritic disease. Autoimmunity 2003, 36:205–210. Jambou F, Zhang W, Menestrier M, et al.: Circulating regulatory anti-T cell receptor antibodies in patients with myasthenia gravis. J Clin Invest 2003, 112:265–274. Jinnin M, Ihn H, Asano Y, et al.: Effect of D-penicillamine on pulmonary fibrosis in patients with systemic sclerosis. Ann Rheum Dis 2003, 62: 1019–1020. Jinnin M, Ihn H, Yazawa N, et al.: Elevated serum levels of manganese superoxide dismutase in patients with eosinophilic fasciitis. Clin Rheumatol 2003, 22:505. Johnson TS, ElKoraie AF, Skill NJ, et al.: Tissue transglutaminase and the progression of human renal scarring. J Am Soc Nephrol 2003, 14:2052–2062. Jorgensen TN, Gubbels MR, Kotzin BL: Links between type I interferons and the genetic basis of disease in mouse lupus. Autoimmunity 2003, 36:491–502. Juarez M, Misischia R, Alarcon GS: Infections in systemic connective tissue diseases: systemic lupus erythematosus, scleroderma, and polymyositis/dermatomyositis. Rheum Dis Clin North Am 2003, 29:163–184. Kagami S, Kakinuma T, Saeki H, et al.: Significant elevation of serum levels of eotaxin-3/CCL26, but not of eotaxin-2/CCL24, in patients with atopic dermatitis: serum eotaxin-3/CCL26 levels reflect the disease activity of atopic dermatitis. Clin Exp Immunol 2003, 134:309–313. Kahaleh B, Meyer O, Scorza R: Assessment of vascular involvement. Clin Exp Rheumatol 2003, 21:S9–S14. Kalluri R, Neilson EG: Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 2003, 112:1776–1784. Kamal SM, Graham CS, He Q, et al.: Kinetics of intrahepatic hepatitis C virus (HCV)-specific CD4(+) T cell responses in HCV and Schistosoma mansoni coinfection: Relation to progression of liver fibrosis. J Infect Dis 2004, 189:1140–1150. Kamali S, Kasapoglu E, Akturk F, et al.: Eosinophilia and hyperimmunoglobulinemia E as the presenting manifestations of Wegener’s granulomatosis. Clin Rheumatol 2003, 22:333–335. Kamei H: Intracellular localization of histidyl-tRNA synthetase/Jo-1 antigen in T24 cells and some other cells. J Autoimmun 2004, 22:201–210. Kasapcopur O, Ozkan HC, Tuysuz B: Linear scleroderma en Coup de Sabre and brain calcification: Is there a pathogenic relationship? J Rheumatol 2003, 30:2724–2725. Kastelein JJP, deGroot E, Sankatsing R: Atherosclerosis measured by B-mode ultrasonography: Effect of statin therapy on disease progression. Am J Med 2004, 116:31–36. Kato T, Miyakawa H, Ishibashi M: Frequency and significance of anti-glutathione S-transferase autoantibody (Anti-GST A1-1) in autoimmune hepatitis. J Autoimmun 2004, 22:211–216. Kawabe S, Abe T, Kawamura H, et al.: Generation of B220(Low)B cells and production of autoantibodies in mice with experimental amyloidosis: association of primordial T cells with this phenomenon. Clin Exp Immunol 2004, 135: 200–208. Kawai K, Saijo K, Oikawa T, et al.: Clinical course and immune response of a renal cell carcinoma patient to adoptive transfer of autologous cytotoxic T lymphocytes. Clin Exp Immunol 2003, 134:264–269. Keogh KA, Specks U: Churg-Strauss syndrome: Clinical presentation, antineutrophil cytoplasmic antibodies, and leukotriene receptor antagonists. Am J Med 2003, 115:284–290. Kerkar N, Choudhuri K, Ma Y, et al.: Cytochrome P4502D6193-212 - A new immunodominant epitope and target of virus/self cross-reactivity in liver kidney microsomal autoantibody type 1-positive liver disease. J Immunol 2003, 170: 1481–1489. Kertai MD, Boersma E, Westerhout CM, et al.: Association between long-term statin use and mortality after successful abdominal aortic aneurysm surgery. Am J Med 2004, 116:96– 103. Khan S, Orenstein S, DiLorenzo C, et al.: Eosinophilic esophagitis: strictures, impactions, dysphagia. Dig Dis Sci 2003, 48:22–29. Khosrotehrani K, Johnson KL, Lau J, et al.: The influence of fetal loss on the presence of fetal cell microchimerism - A systematic review. Arthritis Rheum 2003, 48:3237–3241. Kifor O, McElduff A, Leboff MS, et al.: Activating antibodies to the calcium-sensing receptor in two patients with autoimmune hypoparathyroidism. J Clin Endocrinol Metab 2004, 89: 548–556. Kim LH, Korn JH: Update in systemic sclerosis. Arthritis Rheum Arthritis Care Res 2003, 49: 605–613. KimSaijo M, Akamizu T, Ikuta K, et al.: Generation of a transgenic animal model of hyperthyroid Graves’ disease. Eur J Immunol 2003, 33:2531–2538. Kirman I, Jenkins D, Fowler R, et al.: Naturally occurring antibodies to epithelial cell adhesion molecule (EpCAM). Dig Dis Sci 2003, 48: 2306–2309. Kitani A, Fuss I, Nakamura K, et al.: Transforming growth factor (TGF)-beta 1-producing regulatory T cells induce Smad-mediated interleukin 10 secretion that facilitates coordinated immunoregulatory activity and amelioration of TGFbeta 1-mediated fibrosis. J Exp Med 2003, 198:1179–1188. Kizer JR, Zisman DA, Blumenthal NP, et al.: Association between pulmonary fibrosis and coronary artery disease. Arch Intern Med 2004, 164:551–556. Klein S: The human cost of the SSC. Science 2003, 302:1893. Klion AD, Noel P, Akin C, et al.: Elevated serum tryptase levels identify a subset of patients with a myeloproliferative variant of idiopathic hypereosinophilic syndrome associated with tissue fibrosis, poor prognosis, and imatinib responsiveness. Blood 2003, 101:4660–4666. Kobayashi T, Tanaka S, Okubo M, et al.: Unique epitopes of glutamic acid decarboxylase autoantibodies in slowly progressive type 1 diabetes. J Clin Endocrinol Metab 2003, 88:4768– 4775. Kochhar R, Umbreit JN: Fatal exacerbation of paraneoplastic systemic sclerosis after neoadjuvant chemotherapy in a breast cancer patient. Clin Rheumatol 2004, 23:269–271. Kohn LD, Harii N: Thyrotropin receptor autoantibodies (TSHRAbs): Epitopes, origins and clinical significance. Autoimmunity 2003, 36:331– 337. Kong PL, Zhu TP, Madaio MP, et al.: Role of the H-2 haplotype in Fas-intact lupus-prone MRL mice: association with autoantibodies but not renal disease. Arthritis Rheum 2003, 48:2992–2995. Koury MJ, Newman JH, Murray JJ: Reversal of hypereosinophilic syndrome and lymphomatoid papulosis with mepolizumab and imatinib. Am J Med 2003, 115:587–589. Kovacs L, Papos M, Takacs R, et al.: Autonomic nervous system dysfunction involving the gastrointestinal and the urinary tracts in primary Sjogren’s syndrome. Clin Exp Rheumatol 2003, 21:697–703. KowalBielecka O, Distler O, Kowal K, et al.: Elevated levels of leukotriene B-4 and leukotriene E-4 in bronchoalveolar lavage fluid from patients with scleroderma lung disease. Arthritis Rheum 2003, 48:1639–1646. Kraus VB, Stabler T, Le ET, et al.: Urinary type II collagen neoepitope as an outcome measure for relapsing polychondritis. Arthritis Rheum 2003, 48:2942–2948. Kreisberg RA, Oberman A: Medical management of hyperlipidemia/dyslipidemia. J Clin Endocrinol Metab 2003, 88:2445–2461. Kubo M, CzuwaraLadykowska J, Moussa O, et al.: Persistent down-regulation of Fli1, a suppressor of collagen transcription, in fibrotic scleroderma skin. Am J Pathol 2003, 163:571–581. Kucera P, Novakova D, Behanova M, et al.: Gliadin, endomysial and thyroid antibodies in patients with latent autoimmune diabetes of adults (LADA). Clin Exp Immunol 2003, 133:139– 143. Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Miscellaneous 775 Kuwana M, Sato S, Kikuchi K, et al.: Evaluation of functional disability using the health assessment questionnaire in Japanese patients with systemic sclerosis. J Rheumatol 2003, 30: 1253–1258. Kvasnicka HM, Thiele J, Staib P, et al.: Reversal of bone marrow angiogenesis in chronic myeloid leukemia following imatinib mesylate (STI571) therapy. Blood 2004, 103:3549–3551. Kwak JJ, Chang JE, Lee J, et al.: Chronic eosinophilic pneumonia associated with an initiation of rheumatoid arthritis. Clin Rheumatol 2003, 22:240–243. LaMontagna G, Meli R, Criscuolo T, et al.: Bioactivity of prolactin in systemic sclerosis. Clin Exp Rheumatol 2004, 22:145–150. LaMontagna G, DAngelo S, Valentini G: Crosssectional evaluation of YKL-40 serum concentrations in patients with systemic sclerosis. Relationship with clinical and serological aspects of disease. J Rheumatol 2003, 30:2147–2151. Lamkhioued B, Abdelilah SG, Hamid Q, et al.: The CCR3 receptor is involved in eosinophil differentiation and is up-regulated by Th2 cytokines in CD34+ progenitor cells. J Immunol 2003, 170:537–547. Lapierre P, Johanet C, Alvarez F: Characterization of the B cell response of patients with anti-liver cytosol autoantibodies in type 2 autoimmune hepatitis. Eur J Immunol 2003, 33:1869–1878. Lard LR, vanGaalen FA, Schonkeren JJM, et al.: Association of the-2849 interleukin-10 promoter polymorphism with autoantibody production and joint destruction in rheumatoid arthritis. Arthritis Rheum 2003, 48:1841–1848. Latham KA, Zamora A, Drought H, et al.: Estradiol treatment redirects the isotype of the autoantibody response and prevents the development of autoimmune arthritis. J Immunol 2003, 171: 5820–5827. Layland LE, Wulferink M, Dierkes S, et al.: Druginduced autoantibody formation in mice: triggering by primed CD4(+)CD25(−) T cells, prevention by primed CD4(+)CD25(+) T cells. Eur J Immunol 2004, 34:36–46. Lee DM, Schur PH: Clinical utility of the anti-CCP assay in patients with rheumatic diseases. Ann Rheum Dis 2003, 62:870–874. Lee KH, Chung HS, Kim HS, et al.: Human alphaenolase from endothelial cells as a target antigen of anti-endothelial cell antibody in Behcet’s disease. Arthritis Rheum 2003, 48:2025–2035. Lee CH, Shim JC, Lee YW: Tuberculous fasciitis in scleroderma. Clin Rheumatol 2004, 23:66– 68. Leppilahti M, Tammela TLJ, Huhtala H, et al.: Interstitial cystitis-like urinary symptoms among patients with Sjogren’s syndrome: A population-based study in Finland. Am J Med 2003, 115:62–65. Li YJ, Yang JW, Dai CS, et al.: Role for integrinlinked kinase in mediating tubular epithelial to mesenchymal transition and renal interstitial fibrogenesis. J Clin Invest 2003, 112:503– 516. Liang ZY, Xie C, Chen C, et al.: Pathogenic profiles and molecular signatures of antinuclear autoantibodies rescued from NZM2410 lupus mice. J Exp Med 2004, 199:381–398. Lily O, Palace J, Vincent A: Serum autoantibodies to cell surface determinants in multiple sclerosis: a flow cytometric study. Brain 2004, 127: 269–279. Lim SY, Ghosh SK: Autoreactive responses to an environmental factor. 2. Phthalate-induced anti-DNA specificity is downregulated by autoreactive cytotoxic T cells. Immunology 2004, 112:94–104. Lim SY, Ghosh SK: Autoreactive responses to an environmental factor: 1. phthalate induces anti- bodies exhibiting anti-DNA specificity. Immunology 2003, 110:482–492. Liu YH: Epithelial to mesenchymal transition in renal fibrogenesis: Pathologic significance, molecular mechanism, and therapeutic intervention. J Am Soc Nephrol 2004, 15:1–12. Liu BC, Sun J, Chen Q, et al.: Role of connective tissue growth factor in mediating hypertrophy of human proximal tubular cells induced by angiotensin II. Am J Nephrol 2003, 23:429– 437. Loizou S, Singh S, Wypkerna E, et al.: Anticardiolipin, anti-beta(2)-glycoprotein I and antiprothrombin antibodies in black South African patients with infectious disease. Ann Rheum Dis 2003, 62:1106–1111. Loria P, Lonardo A, Leonardi F, et al.: Non-organspecific autoantibodies in nonalcoholic fatty liver disease: Prevalence and correlates. Dig Dis Sci 2003, 48:2173–2181. Losanoff JE, Sauter ER: Eosinophilic gastroenteritis: Review of the literature. Dig Dis Sci 2003, 48:2214. Lundy SK, Berlin AA, Lukacs NW: Interleukin-12independent down-modulation of cockroach antigen-induced asthma in mice by intranasal exposure to bacterial lipopolysaccharide. Am J Pathol 2003, 163:1961–1968. Luzina IG, Atamas SP, Wise R, et al.: Occurrence of an activated, profibrotic pattern of gene expression in lung CD8+T cells from scleroderma patients. Arthritis Rheum 2003, 48:2262– 2274. Maeshima E, Nishimoto T, Yamashita M, et al.: Progressive systemic sclerosis-polymyositis overlap syndrome with eosinophilic pleural effusion. Rheumatol Int 2003, 23:252–254. Mahler E, Hoebeke J, Levin MJ: Structural and functional complexity of the humoral response against the Trypanosoma cruzi ribosomal P2 beta protein in patients with chronic Chagas’ heart disease. Clin Exp Immunol 2004, 136: 527–534. Maiya S, Desai M, Baruah A, et al.: Cough plate versus cough swab in patients with cystic fibrosis; a pilot study. Arch Dis Child 2004, 89: 577–579. Majeski EI, Paintlia MK, Lopez AD, et al.: Respiratory reovirus 1/L induction of intraluminal fibrosis, a model of bronchiolitis obliterans organizing pneumonia, is dependent on T lymphocytes. Am J Pathol 2003, 163:1467– 1479. Manavalan SJ, Valiando JR, Reeves WH, et al.: Genomic absence of the gene encoding T cell receptor V(Beta)7.2 is linked to the presence of autoantibodies in Sjogren’s syndrome. Arthritis Rheum 2004, 50:187–198. Manganelli P, Fietta P, Martella EM, et al.: Clinical and histological coexistence of inflammatory pseudotumour of the lymph nodes and rheumatoid arthritis. Clin Rheumatol 2003, 22:467–471. Marcolongo R, Tavolini IM, Laveder F, et al.: Immunosuppressive therapy for idiopathic retroperitoneal fibrosis: A retrospective analysis of 26 cases. Am J Med 2004, 116:194–197. MartinMartin LS, Latini A, Pagano A, et al.: A new mathematical model based on clinical and laboratory variables for the diagnosis of Sjogren’s syndrome. Clin Rheumatol 2003, 22: 123–126. Massabki PS, Silva NP, Lourenco DM, et al.: Neuron specific enolase concentration is increased in serum and decreased in platelets of patients with active systemic sclerosis. J Rheumatol 2003, 30:2606–2612. Matsui K, NagyBojarsky K, Laakkonen P, et al.: Lymphatic microvessels in the rat remnant kidney model of renal fibrosis: Aminopeptidase P and podoplanin are discriminatory markers for endothelial cells of blood and lymphatic vessels. J Am Soc Nephrol 2003, 14:1981–1989. Matsuura E, Ohta A, Kanegae F, et al.: Frequency and analysis of factors closely associated with the development of depressive symptoms in patients with scleroderma. J Rheumatol 2003, 30:1782–1787. Mayes MD, Lacey JV, BeebeDimmer J, et al.: Prevalence, incidence, survival, and disease characteristics of systemic sclerosis in a large US population. Arthritis Rheum 2003, 48:2246– 2255. McHugh NJ, Distler O, Giacomelli R, et al.: Non organ based laboratory markers in systemic sclerosis. Clin Exp Rheumatol 2003, 21:S32– S38. McMillan R, Wang L, Tomer A, et al.: Suppression of in vitro megakaryocyte production by antiplatelet autoantibodies from adult patients with chronic ITP. Blood 2004, 103:1364–1369. Meffre E, Schaefer A, Wardemann H, et al.: Surrogate light chain expressing human peripheral B cells produce self-reactive antibodies. J Exp Med 2004, 199:145–150. MeiZahav M, Solomon M, Kawamura A, et al.: Cystic fibrosis presenting as kwashiorkor in a Sri Lankan infant. Arch Dis Child 2003, 88:724– 725. Merkel PA, Clements PJ, Reveille JD, et al.: Current status of outcome measure development for clinical trials in systemic sclerosis. Report from OMERACT 6. J Rheumatol 2003, 30:1630– 1647. Mervis J, Seife C: 10 years after the SSC - Lots of reasons, but few lessons. Science 2003, 302: 38–40. Mervis J: 10 years after the SSC - Scientists are long gone, but bitter memories remain. Science 2003, 302:40–41. Minich WB, Lenzner C, Bergmann A, et al.: A coated tube assay for the detection of blocking thyrotropin receptor autoantibodies. J Clin Endocrinol Metab 2004, 89:352–356. Minich WB, Lenzner C, Morgenthaler NG: Antibodies to TSH-receptor in thyroid autoimmune disease interact with monoclonal antibodies whose epitopes are broadly distributed on the receptor. Clin Exp Immunol 2004, 136:129– 136. Miyachi K, Hirano Y, Horigome T, et al.: Autoantibodies from primary biliary cirrhosis patients with anti-p95c antibodies bind to recombinant p97/VCP and inhibit in vitro nuclear envelope assembly. Clin Exp Immunol 2004, 136:568– 573. Mohan C, Xie C, Liang ZY, et al.: Use of novel elution regimens of autoantibodies in lupus kidneys: comment on the article by Xie et al - Reply. Arthritis Rheum 2004, 50:1700. Mongini PKA, Jackson AE, Tolani S, et al.: Role of complement-binding CD21/CD19/CD81 in enhancing human B cell protection from fasmediated apoptosis. J Immunol 2003, 171: 5244–5254. Moodley YP, Scaffidi AK, Misso NL, et al.: Fibroblasts isolated from normal lungs and those with idiopathic pulmonary fibrosis differ in interleukin-6/gp130-mediated cell signaling and proliferation. Am J Pathol 2003, 163:345– 354. Moore TL, Lunt M, McManus B, et al.: Seventeenpoint dermal ultrasound scoring system - a reliable measure of skin thickness in patients with systemic sclerosis. Rheumatology 2003, 42:1559–1563. Moss RB: Lymphocytes in cystic fibrosis lung disease: a tale of two immunities. Clin Exp Immunol 2004, 135:358–360. Mountantonakis SE, Sakkas LI, Papadopoulos D, et al.: Systemic sclerosis with bilateral hilar adenopathy. Rheumatology 2003, 42:U6–U7. Mpofu S, Rhodes JM, Mpofu CMA, et al.: An unusual cause of acute renal failure in systemic sclerosis. Ann Rheum Dis 2003, 62:1133– 1134. Mrena S, Savola K, Kulmala P, et al.: Genetic modification of risk assessment based on staging of preclinical type 1 diabetes in siblings of af- 776 Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Miscellaneous fected children. J Clin Endocrinol Metab 2003, 88:2682–2689. Murashima A, Fukazawa T, Hirashima M, et al.: Long term prognosis of children born to lupus patients. Ann Rheum Dis 2004, 63:50–53. Muratori P, Muratori L, Gershwin ME, et al.: ‘True’ antimitochondrial antibody-negative primary biliary cirrhosis, low sensitivity of the routine assays, or both? Clin Exp Immunol 2004, 135: 154–158. Murphy E, Freaney R, Bresnihan B, et al.: Increased bone resorption and failure to respond to antiresorptive therapy in progressive dystrophic calcification. Calcif Tissue Int 2003, 73:433– 440. Myou S, Leff AR, Myo S, et al.: Blockade of inflammation and airway hyperresponsiveness in immune-sensitized mice by dominant-negative phosphoinositide 3-kinase-TAT. J Exp Med 2003, 198:1573–1582. Nagayama Y, Mizuguchi H, Hayakawa T, et al.: Prevention of autoantibody mediated Graves’-like hyperthyroidism in mice by interleukin-4, a Th2 cytokine. J Immunol 2003, 170:3522–3527. Nakamura A, Funatomi H, Katagiri A, et al.: A case of autoimmune pancreatitis complicated with immune thrombocytopenia during maintenance therapy with prednisolone. Dig Dis Sci 2003, 48:1968–1971. Nassenstein C, Braun A, Erpenbeck VJ, et al.: The neurotrophins nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4 are survival and activation factors for eosinophils in patients with allergic bronchial asthma. J Exp Med 2003, 198:455– 467. Nielen MMJ, vanSchaardenburg D, Reesink HW, et al.: Specific autoantibodies precede the symptoms of rheumatoid arthritis - A study of serial measurements in blood donors. Arthritis Rheum 2004, 50:380–386. Nielsen CH, Hegedus L, Leslie RGQ: Autoantibodies in autoimmune thyroid disease promote immune complex formation with self antigens and increase B cell and CD4(+) T cell proliferation in response to self antigens. Eur J Immunol 2004, 34:263–272. Nietert PJ, Silver RM: Patterns of hospital, admissions and emergency room visits among patients with scleroderma in South Carolina, USA. J Rheumatol 2003, 30:1238–1243. Nightingale J, Patel S, Suzuki N, et al.: Oncostatin M, a cytokine released by activated mononuclear cells, induces epithelial cell-myofibroblast transdifferentiation via Jak/Stat pathway activation. J Am Soc Nephrol 2004, 15:21– 32. Nishiyama S, Miyawaki S: Splenic marginal zone B-cell lymphoma associated with primary Sjogren’s syndrome. Clin Rheumatol 2004, 23: 242–245. Noble A, Leggat JA, Inderberg EM: CD8(+) immunoregulatory cells in the graft-versus-host reaction: CD8 T cells activate dendritic cells to secrete interleukin-12/interleukin-18 and induce T helper I autoantibody. Immunology 2003, 109:476–486. Nozawa K, Fritzler MJ, vonMuhlen CA, et al.: Giantin is the major Golgi autoantigen in human antiGolgi complex sera. Arthritis Res Ther 2004, 6:R95–R102. Nutku E, Aizawa H, Hudson SA, et al.: Ligation of Siglec-8: a selective mechanism for induction of human eosinophil apoptosis. Blood 2003, 101:5014–5020. Oakley F, Trim N, Constandinou CM, et al.: Hepatocytes express nerve growth factor during liver injury - Evidence for paracrine regulation of hepatic stellate cell apoptosis. Am J Pathol 2003, 163:1849–1858. Oba Y, Chung HY, Choi SJ, et al.: Eosinophil chemotactic factor-1 (ECF-L): A novel osteoclast stimulating factor. J Bone Miner Res 2003, 18:1332–1341. Ogilvie P, Paoletti S, ClarkLewis I, et al.: Eotaxin-3 is a natural antagonist for CCR2 and exerts a repulsive effect on human monocytes. Blood 2003, 102:789–794. Okon K, Szumera A, Kuzniewski M: Are CD34+ cells found in renal interstitial fibrosis? Am J Nephrol 2003, 23:409–414. OstrandRosenberg S: Animal models of tumor immunity, immunotherapy and cancer vaccines. Curr Opin Immunol 2004, 16:143–150. Ott PA, Dittrich MT, Herzog BA, et al.: T cells recognize multiple GAD65 and proinsulin Epitopes in human type 1 diabetes, suggesting determinant spreading. J Clin Immunol 2004, 24:327–339. Ou Y, Enarson P, Rattner JB, et al.: The nuclear pore complex protein Tpr is a common autoantigen in sera that demonstrate nuclear envelope staining by indirect immunofluorescence. Clin Exp Immunol 2004, 136:379–387. Pagnotta A, Specchia N, Soccetti A, et al.: Responsiveness of Dupuytren’s disease fibroblasts to 5 alpha-dihydrotestosterone. J Hand Surg [Am] 2003, 28A:1029–1034. Pamuk ON, Pamuk GE, Turgut B, et al.: Scleroderma in a patient with X-linked agammaglobulinaemia. Scand J Rheumatol 2004, 33:59–60. Papamattheou MG, Routsias JG, Karagouni EE, et al.: T cell help is required to induce idiotypicanti-idiotypic autoantibody network after immunization with complementary epitope 289308aa of La/SSB autoantigen in nonautoimmune mice. Clin Exp Immunol 2004, 135:416–426. Park SH, Silva M: Effect of intermittent pneumatic soft-tissue compression on fracture-healing in an animal model. J Bone Joint Surg (Am) 2003, 85A:1446–1453. Pearce SHS, Leech NJ: Editorial: Toward precise forecasting of autoimmune endocrinopathy. J Clin Endocrinol Metab 2004, 89:544–547. Pearson JE, Silman AJ: Risk of cancer in patients with scleroderma. Ann Rheum Dis 2003, 62: 697–699. Pellicano R, Touscoz GA, Smedile A, et al.: Prevalence of non-organ-specific autoantibodies in patients suffering from duodenal ulcer with and without Helicobacter pylori infection. Dig Dis Sci 2004, 49:395–398. Phillips RM, Stubbs VEL, Henson MR, et al.: Variations in eosinophil chemokine responses: An investigation of CCR1 and CCR3 function, expression in atopy, and identification of a functional CCR1 promoter. J Immunol 2003, 170:6190–6201. Pichurin PN, Chen CR, Nagayama Y, et al.: Evidence that factors other than particular thyrotropin receptor T cell epitopes contribute to the development of hyperthyroidism in murine Graves’ disease. Clin Exp Immunol 2004, 135: 391–397. Pichurin P, Aliesky H, Chen CR, et al.: Thyrotrophin receptor-specific memory T cell responses require normal B cells in a murine model of Graves’ disease. Clin Exp Immunol 2003, 134: 396–402. Pierangeli SS, Blank M, Liu XW, et al.: A peptide that shares similarity with bacterial antigens reverses thrombogenic properties of antiphospholipid antibodies in vivo. J Autoimmun 2004, 22:217–225. Piliponsky AM, Gleich GJ, Nagler A, et al.: Non-IgE dependent activation of human lung and cord blood-derived mast cells is induced by eosinophil major basic protein and modulated by the membrane form of stem cell factor. Blood 2003, 101:1898–1904. Pishko EJ, Kirimanjeswara GS, Pilione MR, et al.: Anti body-mediated bacterial clearance from the lower respiratory tract of mice requires complement component C3. Eur J Immunol 2004, 34:184–193. Pittoni V, Sorice M, Circella A, et al.: Specificity of antinuclear and antiphospholipid antibodies in sera of patients with autoimmune lymphoprolif- erative disease (ALD). Clin Exp Rheumatol 2003, 21:377–385. Poole JL, Brower L: Validity of the scleroderma functional assessment questionnaire. J Rheumatol 2004, 31:402–403. Pugazhenthi M, Cooper D, Ratnakant BS, et al.: Hypercapnic respiratory failure in systemic sclerosis. JCR - J Clin Rheumatol 2003, 9:43– 46. Raghu G: The role of gastroesophageal reflux in idiopathic pulmonary fibrosis. Am J Med 2003, 115:60S–64S. Rand JH, Wu XX, Quinn AS, et al.: Human monoclonal antiphospholipid antibodies disrupt the annexin A5 anticoagulant crystal shield on phospholipid bilayers - Evidence from atomic force microscopy and functional assay. Am J Pathol 2003, 163:1193–1200. Reiners KS, Hansen HP, Krussmann A, et al.: Selective killing of B-cell hybridomas targeting proteinase 3, Wegener’s autoantigen. Immunology 2004, 112:228–236. Reumaux D, Duthilleul P, Roos D: Pathogenesis of diseases associated with antineutrophil cytoplasm autoantibodies. Hum Immunol 2004, 65:1–12. Riccieri V, Rinaldi T, Spadaro A, et al.: Interleukin13 in systemic sclerosis: relationship to nailfold capillaroscopy abnormalities. Clin Rheumatol 2003, 22:102–106. Rice NA, Leinwand LA: Skeletal myosin heavy chain function in cultured lung myofibroblasts. J Cell Biol 2003, 163:119–129. Richards HL, Herrick AL, Griffin K, et al.: Systemic sclerosis: Patients’ perceptions of their condition. Arthritis Rheum Arthritis Care Res 2003, 49:689–696. Riente L, Bongiorni F, Nacci A, et al.: Antibodies to inner ear antigens in Meniere’s disease. Clin Exp Immunol 2004, 135:159–163. Ringer AJ, Guterman LR, Hopkins LN: Site-specific thromboembolism: A novel animal model for stroke. AJNR Am J Neuroradiol 2004, 25:329–332. Robbins RJ: Statins sentence thyroid cancer cells to death Rho. J Clin Endocrinol Metab 2003, 88:3019–3020. Robinson WH, Utz PJ, Steinman L: Genomic and proteomic analysis of multiple sclerosis - Opinion. Curr Opin Immunol 2003, 15:660–667. Rosenson RS: Current overview of statin-induced myopathy. Am J Med 2004, 116:408–416. Russo MA, Hogenauer C, Coates SW, et al.: Abnormal passive chloride absorption in cystic fibrosis jejunum functionally opposes the classic chloride secretory defect. J Clin Invest 2003, 112:118–125. Sabbatini A, Tacchetti C, Tommasi S, et al.: Use of human chondrocyte cell cultures to identify and characterize reactive antibodies in rheumatoid arthritis sera. Clin Exp Rheumatol 2003, 21:587–592. Saeki T, Fujita N, Kourakata H, et al.: Two cases of hypertrophic pachymeningitis associated with myeloperoxidase antineutrophil cytoplasmic autoantibody (MPO-ANCA)-positive pulmonary silicosis in tunnel workers. Clin Rheumatol 2004, 23:76–80. Salomonsson S, Jonsson MV, Skarstein K, et al.: Cellular basis of ectopic germinal center formation and autoantibody production in the target organ of patients with Sjogren’s syndrome. Arthritis Rheum 2003, 48:3187–3201. Salomonsson S, Ottosson L, Safsten P, et al.: Cloning and characterization of two human Ro52specific monoclonal autoantibodies directed towards a domain associated with congenital heart block. J Autoimmun 2004, 22:167–177. Sandqvist G, Eklund M, Akesson A, et al.: Daily activities and hand function in women with scleroderma. Scand J Rheumatol 2004, 33: 102–107. Santiago M, deCastro DO, Costa CA, et al.: Keloidal scleroderma. Clin Rheumatol 2004, 23: 50–51. Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Miscellaneous 777 Sato S, Ishida W, Takehara K: A case of juvenile systemic sclerosis with disease onset at six months old. Clin Rheumatol 2003, 22:162– 163. Schibler A, vonderHeiden R, Birrer P, et al.: Prospective randomised treatment with recombinant human growth hormone in cystic fibrosis. Arch Dis Child 2003, 88:1078–1081. Schmidt RE, Dorsey DA, Beaudet LN, et al.: Nonobese diabetic mice rapidly develop dramatic sympathetic neuritic dystrophy - A new experimental model of diabetic autonomic neuropathy. Am J Pathol 2003, 163:2077–2091. Schmitt A, Schubert D, Sengler C, et al.: Autoantibodies against glucose-6-phosphate isomerase are not a diagnostic marker for juvenile idiopathic arthritis. Ann Rheum Dis 2004, 63:463. Schultz H, Heintz H, vanZandbergen G, et al.: ANCA against the bactericidal/permeability increasing protein (BPI-ANCA) can compromise the antibiotic function of BPI in a Wegener’s granulomatosis patient. Clin Exp Rheumatol 2003, 21:763–766. Schultz H, Schinke S, Weiss J, et al.: BPI-ANCA in transporter associated with antigen presentation (TAP) deficiency: possible role in susceptibility to Gram-negative bacterial infections. Clin Exp Immunol 2003, 133:252–259. Schumacher HR, Pessler F, Chen LX: Diagnosing early rheumatoid arthritis (RA). What are the problems and opportunities? Clin Exp Rheumatol 2003, 21:S15–S19. Schwarze C, Terjung B, Lilienweiss P, et al.: IgA class antineutrophil cytoplasmic antibodies in primary sclerosing cholangitis and autoimmune hepatitis. Clin Exp Immunol 2003, 133:283– 289. Seetharamaiah GS: Animal models of Graves’ hyperthyroidism. Autoimmunity 2003, 36:381– 387. Sehmi R, Dorman S, Baatjes A, et al.: Allergin-induced fluctuation in CC chemokine receptor 3 expression on bone marrow CD34(+) cells from asthmatic subjects: significance for mobilization of haemopoietic progenitor cells in allergic inflammation. Immunology 2003, 109: 536–546. Seife C: 10 years after the SSC - Physics tries to leave the tunnel. Science 2003, 302:36–38. Sekigawa I, Tokano Y, Yoshiike T, et al.: Relationship between serum IgE and autoantibody levels in SLE patients. Clin Exp Rheumatol 2003, 21:683. Sellge G, Lorentz A, Gebhardt T, et al.: Human intestinal fibroblasts prevent apoptosis in human intestinal mast cells by a mechanism independent of stem cell factor, IL-3, IL-4, and nerve growth factor. J Immunol 2004, 172:260–267. SermetGaudelus I, Souberbielle JC, Azhar I, et al.: Insulin-like growth factor I correlates with lean body mass in cystic fibrosis patients. Arch Dis Child 2003, 88:956–961. Seth D, Leo MA, McGuinness PH, et al.: Gene expression profiling of alcoholic liver disease in the baboon (Papio hamadryas) and human liver. Am J Pathol 2003, 163:2303–2317. Sharma P, Balan V, Hernandez J, et al.: Hepatic steatosis in hepatitis C virus genotype 3 infection: Does it correlate with body mass index, fibrosis, and HCV risk factors? Dig Dis Sci 2004, 49:25–29. Shen CR, Youssef AR, Devine A, et al.: Peptides containing a dominant T-cell epitope from red cell band 3 have in vivo immunomodulatory properties in NZB mice with autoimmune hemolytic anemia. Blood 2003, 102:3800–3806. Silva EEC, Ramos OP, Bauab TM, et al.: Yersinia enterocolitica O:3 isolated from patients with or without reactive arthritis induces polyclonal activation of B cells and autoantibody production in vivo. Autoimmunity 2003, 36:261–268. Silverman GJ, Carson DA: Roles of B cells in rheumatoid arthritis. Arthritis Res Ther 2003, 5:S1–S6. Simms RW, Korn JH: The role of leukotrienes in alveolitis associated with scleroderma. Arthritis Rheum 2003, 48:1478–1480. Singh KP, Gerard HC, Hudson AP, et al.: Expression of matrix metalloproteinases and their inhibitors during the resorption of schistosome egg-induced fibrosis in praziquantel-treated mice. Immunology 2004, 111:343–352. Skoldberg F, Rorsman F, Perheentupa J, et al.: Analysis of antibody reactivity against cysteine sulfinic acid decarboxylase, a pyridoxal phosphate-dependent enzyme, in endocrine autoimmune disease. J Clin Endocrinol Metab 2004, 89:1636–1640. Sokka T, Willoughby J, Yazici Y, et al.: Databases of patients with early rheumatoid arthritis in the USA. Clin Exp Rheumatol 2003, 21:S146– S153. Song RP, Kubo M, Morse D, et al.: Carbon monoxide induces cytoprotection in rat orthotopic lung transplantation via anti-inflammatory and anti-apoptotic effects. Am J Pathol 2003, 163: 231–242. Sood M, Adams JE, Mughal MZ: Lean body mass in children with cystic fibrosis. Arch Dis Child 2003, 88:836. Spencer FA, Allegrone J, Goldberg RJ, et al.: Association of statin therapy with outcomes of acute coronary syndromes: The GRACE Study. Ann Intern Med 2004, 140:857–866. Staikou EV, Routsias JG, Makri AA, et al.: Calreticulin binds preferentially with B cell linear epitopes of Ro60 kD autoantigen, enhancing recognition by anti-Ro60 kD autoantibodies. Clin Exp Immunol 2003, 134:143–150. Stummvoll GH, Aringer M, Grisar J, et al.: Increased transendothelial migration of scleroderma lymphocytes. Ann Rheum Dis 2004, 63:569–574. Sucato DJ, Hedequist D, Zhang H, et al.: Recombinant human bone morphogenetic protein-2 enhances anterior spinal fusion in a thoracoscopically instrumented animal model. J Bone Joint Surg (Am) 2004, 86A:752–762. Sugiura Y, Banno S, Matsumoto Y, et al.: Transforming growth factor beta 1 gene polymorphism in patients with systemic sclerosis. J Rheumatol 2003, 30:1520–1523. Sultan N, Pope JE, Clements PJ: The health assessment questionnaire (HAQ) is strongly predictive of good outcome in early diffuse scleroderma: results from an analysis of two randomized controlled trials in early diffuse scleroderma. Rheumatology 2004, 43:472– 478. Super M: Cystic fibrosis newborn screening and detection of carriers. Arch Dis Child 2003, 88:F448–F449. Swartz RD, Crofford LJ, Phan SH, et al.: Nephrogenic fibrosing dermopathy - A novel cutaneous fibrosing disorder in patients with renal failure. Am J Med 2003, 114:563–572. Takagi K, Kawaguchi Y, Hara M, et al.: Serum nitric oxide (NO) levels in systemic sclerosis patients: correlation between NO levels and clinical features. Clin Exp Immunol 2003, 134: 538–544. Takahashi H, Ebihara S, Kanda A, et al.: Increased susceptibility to airway responses in CD40deficient mice. Clin Exp Immunol 2003, 133: 22–29. Takeshita K, Hayashi M, Iino S, et al.: Increased expression of plasminogen activator inhibitor-1 in cardiomyocytes contributes to cardiac fibrosis after myocardial infarction. Am J Pathol 2004, 164:449–456. Tan EM, Shi FD: Relative paradigms between autoantibodies in lupus and autoantibodies in cancer. Clin Exp Immunol 2003, 134:169–177. Tanko LB, Bagger YZ, Nielsen SB, et al.: Does serum cholesterol contribute to vertebral bone loss in postmenopausal women? Bone 2003, 32:8–14. Taylor JG, Bolster MB: Bronchiolitis, obliterans with organizing pneumonia associated with scleroderma and scleroderma spectrum diseases. JCR - J Clin Rheumatol 2003, 9:239–245. Tazawa H, Okada F, Kobayashi T, et al.: Infiltration of neutrophils is required for acquisition of metastatic phenotype of benign murine fibrosarcoma cells - Implication of inflammationassociated carcinogenesis and tumor progression. Am J Pathol 2003, 163:2221–2232. Theocharis SE, Papadimitriou LJ, Retsou ZP, et al.: Granulocyte-colony stimulating factor administration ameliorates liver regeneration in animal model of fulminant hepatic failure and encephalopathy. Dig Dis Sci 2003, 48:1797–1803. Thrasyvoulides A, Lymberi P: Evidence for intramolecular B-cell epitope spreading during experimental immunization with an immunogenic thyroglobulin peptide. Clin Exp Immunol 2003, 132:401–407. Tiittanen M, Knip M, Vaarala O: Anti-insulin activity in IgG-fractions from children with newly-diagnosed type 1 diabetes and negative for insulin autoantibodies. Autoimmunity 2004, 37:45– 49. Torn C, Gupta M, Zake LN, et al.: Heterozygosity for MICA5.0/MICA5.1 and HLA-DR3DQ2/DR4-DQ8 are independent genetic risk factors for latent autoimmune diabetes in adults. Hum Immunol 2003, 64:902–909. Tusa RJ, Mustari MJ, Das VE, et al.: Animal models for visual deprivation-induced strabismus and nystagmus. Ann N Y Acad Sci 2002, 956: 346–360. Uchida K, Nakata K, Trapnell BC, et al.: High-affinity autoantibodies specifically eliminate granulocyte-macrophage colony-stimulating factor activity in the lungs of patients with idiopathic pulmonary alveolar proteinosis. Blood 2004, 103:1089–1098. Uibu T, Oksa P, Auvinen A, et al.: Asbestos exposure as a risk factor for retroperitoneal fibrosis. Lancet 2004, 363:1422–1426. Ulanet DB, Flavahan NA, CasciolaRosen L, et al.: Selective cleavage of nucleolar autoantigen B23 by granzyme B in differentiated vascular smooth muscle cells - Insights into the association of specific autoantibodies with distinct disease phenotypes. Arthritis Rheum 2004, 50:233–241. Utz PJ, Genovese MC, Robinson WH: Unlocking the “PAD” lock on rheumatoid arthritis. Ann Rheum Dis 2004, 63:330–332. Valentini G, Bencivelli W, Bombardieri S, et al.: European Scleroderma Study Group to define disease activity criteria for systemic sclerosis. III. Assessment of the construct validity of the preliminary activity criteria. Ann Rheum Dis 2003, 62:901–903. Valentini G, DAngelo S, DellaRossa A, et al.: European Scleroderma Study Group to define disease activity criteria for systemic sclerosis. IV. Assessment of skin thickening by modified Rodnan skin score. Ann Rheum Dis 2003, 62: 904–905. Varadi K, Schreiner J, Plaimauer B, et al.: ADAMTS13 autoantibody detection by quantitative immunoblotting. Blood 2003, 102:1932– 1933. Varela M, Garvin JL: Acute and chronic regulation of thick ascending limb endothelial nitric oxide synthase by statins. J Am Soc Nephrol 2004, 15:269–275. Vernon DP: The lesson of the SSC. Science 2003, 302:1893. Vesely R, Vargova V, Ravelli A, et al.: Serum level of KL-6 as a marker of interstitial lung disease in patients with juvenile systemic sclerosis. J Rheumatol 2004, 31:795–800. Virag JI, Murry CE: Myofibroblast and endothelial cell proliferation during murine myocardial infarct repair. Am J Pathol 2003, 163:2433– 2440. Viskari H, Paronen J, Keskinen P, et al.: Humoral beta-cell autoimmunity is rare in patients with 778 Raynaud phenomenon, scleroderma, and other fibrosing syndromes: Miscellaneous the congenital rubella syndrome. Clin Exp Immunol 2003, 133:378–383. VonScheven E, Lu TT, Emery HM, et al.: Thrombosis and pediatric Wegener’s granulomatosis: Acquired and genetic risk factors for hypercoagulability. Arthritis Rheum Arthritis Care Res 2003, 49:862–865. Vossenaar ER, Zendman AJW, vanVenrooij WJ: Citrullination, a possible functional link between susceptibility genes and rheumatoid arthritis. Arthritis Res Ther 2003, 6:1–5. Vourlekis JS, Schwarz MI, Cherniack RM, et al.: The effect of pulmonary fibrosis on survival in patients with hypersensitivity pneumonitis. Am J Med 2004, 116:662–668. Wang HS, Clarke SH: Regulation of B-cell development by antibody specificity. Curr Opin Immunol 2004, 16:246–250. Waters ST, McDuffie M, Bagavant H, et al.: Breaking tolerance to double stranded DNA, nucleosome, and other nuclear antigens is not required for the pathogenesis of lupus glomerulonephritis. J Exp Med 2004, 199:255–264. Weber GF: The absence of CD44 ameliorates Fas(Lpr/lpr) disease. Autoimmunity 2004, 37:1–8. Weerasooriya VS, White FV, Shepherd RW: Hepatic fibrosis and survival in biliary atresia. J Pediatr 2004, 144:123–125. Wernhoff P, Olofsson P, Holmdahl R: The genetic control of rheumatoid factor production in a rat model of rheumatoid arthritis. Arthritis Rheum 2003, 48:3584–3596. White B: Interstitial lung disease in scleroderma. Rheum Dis Clin North Am 2003, 29:371–390. Wilkinson K, Velloso ERP, Lopes LF, et al.: Cloning of the t(L;5)(Q23;q33) in a myeloproliferative disorder associated with eosinophilia: involvement of PDGFRB and response to imatinib. Blood 2003, 102:4187–4190. Williams TJ: The eosinophil enigma. J Clin Invest 2004, 113:507–509. Willison HJ, Townson K, Veitch J, et al.: Synthetic disialylgalactose immunoadsorbents deplete anti-GQ1b antibodies from autoimmune nueropathy sera. Brain 2004, 127:680–691. Woerly G, Honda K, Loyens M, et al.: Peroxisome proliferator-activated receptors alpha and gamma down-regulate allergic inflammation and eosinophil activation. J Exp Med 2003, 198:411–421. Wollina U: Increased risk of cancer in patients with scleroderma: no risk in patients with morphoea? Ann Rheum Dis 2004, 63:611–612. Wu Z, Holwill SDJ, Oliveira DBG: Desferrioxamine modulates chemically induced T helper 2-mediated autoimmunity in the rat. Clin Exp Immunol 2004, 135:194–199. Xie C, Liang ZY, Chang SH, et al.: Use of a novel elution regimen reveals the dominance of polyreactive antinuclear autoantibodies in lupus kidneys. Arthritis Rheum 2003, 48:2343–2352. Xu B, Broome U, Uzunel M, et al.: Capillarization of hepatic sinusoid by liver endothelial cell-reactive autoantibodies in patients with cirrhosis and chronic hepatitis. Am J Pathol 2003, 163: 1275–1289. Yamaguchi A, Katsuyama K, Nagahama K, et al.: Possible role of autoantibodies in the pathophysiology of GM2 gangliosidoses. J Clin Invest 2004, 113:200–208. Yamamoto T: Potential roles of CCL2/monocyte chemoattractant protein-1 in the pathogenesis of cutaneous sclerosis. Clin Exp Rheumatol 2003, 21:369–375. Yamamoto T, Nishioka K: Role of monocyte chemoattractant protein-1 and its receptor, CCR-2, in the pathogenesis of bleomycin-induced scleroderma. J Invest Dermatol 2003, 121:510–516. Yamamoto T, Akisue T, Marui T, et al.: Subcutaneous, intramuscular, and intraosseous synovial cyst formation around the knee in rheumatoid arthritis/systemic lupus erythematosus overlap syndrome. J Rheumatol 2003, 30:1351–1353. Yanaba K, Hasegawa M, Hamaguchi Y, et al.: Longitudinal analysis of serum KL-6 levels in patients with systemic sclerosis: association with the activity of pulmonary fibrosis. Clin Exp Rheumatol 2003, 21:429–436. Yanaba K, Takehara K, Sato S: Serum concentrations of soluble P-selectin glycoprotein ligand-1 are increased in patients with systemic sclerosis: association with lower frequency of pulmonary fibrosis. Ann Rheum Dis 2004, 63: 583–587. Yang J, Shultz RW, Mars WM, et al.: Disruption of tissue type plasminogen activator gene in mice reduces renal interstitial fibrosis in obstructive nephropathy. J Clin Invest 2002, 110:1525– 1538. Yong PFK, Hamour SMA, Burns A: Reversible posterior leukoencephalopathy in a patient with systemic sclerosis/systemic lupus erythematosus overlap syndrome. Nephrol Dial Transplant 2003, 18:2660–2662. Yoshida K, Tsutsumi A, Ohnishi Y, et al.: T cell epitopes of prothrombin in patients with antiphospholipid syndrome. Ann Rheum Dis 2003, 62: 905–906. ZhangHoover J, SteinStreilein J: Tolerogenic APC generate CD8(+) T regulatory cells that modulate pulmonary interstitial fibrosis. J Immunol 2004, 172:178–185. Zhou XD, Tan FK, Wang N, et al.: Genome-wide association study for regions of systemic sclerosis susceptibility in a Choctaw Indian population with high disease prevalence. Arthritis Rheum 2003, 48:2585–2592. Zhu H, Bona C, McGaha TL: Polymorphisms of the TGF-beta 1 promoter in tight skin (TSK) mice. Autoimmunity 2004, 37:51–55. dePerrot M, Young K, Imai Y, et al.: Recipient T cells mediate reperfusion injury after lung transplantation in the rat. J Immunol 2003, 171:4995–5002. deVillartay JP, Poinsignon C, deChasseval R, et al.: Human and animal models of V(D)J recombination deficiency. Curr Opin Immunol 2003, 15:592–598. deWinter RJ, Koch KT, vanStraalen JP, et al.: Creactive protein and coronary events following percutaneous coronary angioplasty. Am J Med 2003, 115:85–90. diGirolamo C, Rengo C, Ferrucci MG, et al.: Systemic sclerosis could mask the presentation of psoriasis in a patient with symptomatic and bilateral sacroiliitis. Scand J Rheumatol 2003, 32:186–188. vanGaalen FA, Toes REM, Ditzel HJ, et al.: Association of autoantibodies to glucose-6-phosphate isomerase with extraarticular complications in rheumatoid arthritis. Arthritis Rheum 2004, 50:395–399. vanHoorn JHL, Hendriks JJE, Vermeer C, et al.: Vitamin K supplementation in cystic fibrosis. Arch Dis Child 2003, 88:974–975. vanLent P, Nabbe KC, Boross P, et al.: The inhibitory receptor Fc gamma RII reduces joint inflammation and destruction in experimental immune complex-mediated arthritides not only by inhibition of Fc gamma RI/III but also by efficient clearance and endocytosis of immune complexes. Am J Pathol 2003, 163:1839– 1848. vanPaassen P, Damoiseaux J, Tervaert JWC: Laboratory assessment in musculoskeletal disorders. Best Pract Res Clin Rheumatol 2003, 17:475–494. 10 years after the SSC - The power of words: The SSC in literature. Science 2003, 302:41. Manual of signs, symptoms, methods and procedures for the assessment of the patient with SSc. Clin Exp Rheumatol 2003, 21:S49–S56. List of journals scanned The Index Medicus abbreviation is given in parentheses. Acta Orthopaedica Scandinavica (Acta Orthop Scand) Advances in Immunology (Adv Immunol) American Journal of Human Genetics (Am J Hum Genet) American Journal of Medicine (Am J Med) American Journal of Nephrology (Am J Nephrol) American Journal of Neuroradiology (AJNR Am J Neuroradiol) American Journal of Pathology (Am J Pathol) American Journal of Physical Medicine and Rehabilitation (Am J Phys Med American Journal of Sports Medicine (Am J Sports Med) Amsterdam: Elsevier (Amsterdam: Elsevier) Annals of Internal Medicine (Ann Intern Med) Annals of the New York Academy of Sciences (Ann N Y Acad Sci) Annals of the Rheumatic Diseases (Ann Rheum Dis) Annual Review of Immunology (Annu Rev Immunol) Archives of Disease in Childhood (Arch Dis Child) Archives of Internal Medicine (Arch Intern Med) Archives of Physical Medicine and Rehabilitation (Arch Phys Med Rehabil) Arthritis and Rheumatism (Arthritis Rheum) Arthritis Care and Research (Arthritis Care Res) Arthritis Research (Arthritis Res) Autoimmunity (Autoimmunity) Journal Journal Journal Journal Journal Journal Journal Journal Journal Journal Journal Journal Journal Journal Journal Journal Journal Journal Journal Baillieres Clinical Rheumatology (Baillieres Clin Rheumatol) Best Practice and Research in Clinical Rheumatology (Best Pract Res Clin Blood (Blood) Bone (Bone) Brain (Brain) British Medical Journal (BMJ) Bulletin on the Rheumatic Diseases (Bull Rheum Dis) Lancet (Lancet) Lupus (Lupus) Calcified Tissue International (Calcif Tissue Int) Clinical and Experimental Immunology (Clin Exp Immunol) Clinical and Experimental Rheumatology (Clin Exp Rheumatol) Clinical Immunology (Clin Immunol) Clinical Infectious Diseases (Clin Infect Dis) Clinical Orthopaedics and Related Research (Clin Orthop) Clinical Rheumatology (Clin Rheumatol) Clinics in Sports Medicine (Clin Sports Med) Curr Rheumatol Rep (Curr Rheumatol Rep) Current Opinion in Immunology (Curr Opin Immunol) Current Opinion in Orthopedics (Curr Opin Orthop) Current Opinion in Rheumatology (Curr Opin Rheumatol) Digestive Diseases and Sciences (Dig Dis Sci) Eur]) European Journal of Immunology (Eur J Immunol) Human Immunology (Hum Immunol) Human Molecular Genetics (Hum Mol Genet) Immunology (Immunology) JAMA Journal of the American Medical Association (JAMA) JCR - Journal of Clinical Rheumatology (JCR - J Clin Rheumatol) Jmri - Journal of Magnetic Resonance Imaging (J Magn Reson Imaging) Journal of Arthroplasty (J Arthroplasty) Journal of Autoimmunity (J Autoimmun) Journal of Biological Chemistry (J Biol Chem) Journal of Bone and Joint Surgery American Volume (J Bone Joint Surg (Am)) Journal of Bone and Joint Surgery British Volume (J Bone Joint Surg (Br)) ISSN 1040–8711 © 2004 Lippincott Williams & Wilkins of of of of of of of of of of of of of of of of of of of Bone and Mineral Research (J Bone Miner Res) Cell Biology (J Cell Biol) Clinical Endocrinology and Metabolism (J Clin Endocrinol Metab) Clinical Immunology (J Clin Immunol) Clinical Investigation (J Clin Invest) Experimental Medicine (J Exp Med) Hand Surgery American Volume (J Hand Surg [Am]) Hand Surgery British and European Volume (J Hand Surg [Br and Immunology (J Immunol) Infectious Diseases (J Infect Dis) Investigative Dermatology (J Invest Dermatol) Medical Genetics (J Med Genet) Neuropathology and Experimental Neurology (J Neuropathol Exp Orthopaedic Rheumatology (J Orthop Rheumatol) Pediatric Orthopedics (J Pediatr Orthop) Pediatrics (J Pediatr) Rheumatology (J Rheumatol) the American Society of Nephrology (J Am Soc Nephrol) the Royal Society of Medicine (J R Soc Med) Kidney International (Kidney Int) Medical Care (Med Care) Medicine (Medicine) Micros Res Tech (Micros Res Tech) Nature (Nature) Nature Medicine (Nat Med) Nephrology, Dialysis, and Transplantation (Nephrol Dial Transplant) Neurol) Neurology (Neurology) Neuroscience Letters (Neurosci Lett) New England Journal of Medicine (N Engl J Med) Occupational Medicine (Occup Med) Osteoarthritis and Cartilage (Osteoarth Cartilage) Osteoarthritis Cartilage (Osteoarthritis Cartilage) Osteoporosis International (Osteoporosis Int) Pain (Pain) Pediatrics (Pediatrics) Proceedings of the Association of American Physicians (Proc Assoc Am Phys) Proceedings of the National Academy of Sciences of the United States of America (Proc Natl Acad Sci USA) Radiology (Radiology) Rehabil) Rheumatic Disease Clinics of North America (Rheum Dis Clin North Am) Rheumatol) Rheumatology (Rheumatology) Rheumatology International (Rheumatol Int) Scandinavian Journal of Rheumatology (Scand J Rheumatol) Science (Science) Seminars in Arthritis and Rheumatism (Semin Arthritis Rheum) Seminars in Neurology (Semin Neurol) Spine (Spine) Zeitschrift für Rheumatologie (Z Rheumatol) Index to authors Volume 16 2004 Abramson SB, 199 Abril A, 51 Anandarajah AP, 338 Anolik J, 180, 505 Arakaki R, 522 Arce E, 577 Askling J, 254 Atzeni F, 287 Backman CL, 148 Baecklund E, 254 Baldock PA, 450 Bardin T, 76 Benseler S, 43 Berenbaum F, 616 Bhatt DL, 18 Brune K, 628 Buckwalter JA, 634 Burmester GR, 246 Buxbaum JN, 67 Calabrese LH, 18 Callan MFC, 399 Cepeda EJ, 723 Chamian F, 331 Chinoy H, 707 Christner PJ, 746 Christopher-Stine L, 700 Cohen MD, 51 Conaghan PG, 435 Conn DL, 192 Cooper RG, 707 Cortinovis D, 357 Costello JC, 268 Crow MK, 541 Dechant SA, 177 deHueck A, 138 Delmas PD, 428 Dörner T, 246 Duffy CM, 102 Egerer K, 246 Eisman JA, 450 Ekbom A, 254 Emery P, 435 Fahmi H, 623 Feist E, 246 Firestein GS, 231 Fitzgerald GK, 143 Garnero P, 428 Gaston JSH, 371 Gay RE, 238 Gay S, 238, 411 Gershwin ME, 406 Ginzler EM, 499 Gladman DD, 329 Gohr C, 263 Goronzy JJ, 212 Gowans SE, 138 Gravallese EM, 419 Häkkinen A, 132 Hamilton CD, 393 Hamuryudan V, 38 Hanna MG, 684 Harley JB, 534 Hashkes PJ, 560 Hayashi Y, 522 Heeringa P, 4 Helliwell PS, 344 Hengstman GJD, 692 Hinz B, 628 Hirsch R, 553 Holton JL, 684 Hootman JM, 119 Horowitz MC, 464 Hungerford DS, 443 Huugen D, 4 Imperato AK, 199 Isenberg D, 665 Ishimaru N, 522 Jarrett S, 435 Jimenez SA, 746 Jones LC, 443 Jordan JM, 62 Kahaleh MB, 718 Kanangat S, 733 Kato T, 604 Kelley JT, III, 192 Kent PD, 56 Kietz D, 555 Kingsley GH, 678 Kirou KA, 541 Kirwan JR, 125 Kissin EY, 31 Klareskog L, 254 Köhler L, 380 Krueger JG, 331 Kuipers J, 380 Kyburz D, 411 Kyttaris VC, 548 Lane NE, 457 Langford CA, 1 Lauener RP, 411 Leveille SG, 114 Linn ML, 374 Liu Y, 357 Looney RJ, 180 Lorenzo JA, 464 Luthra HS, 56 Luyten FP, 599 Maksimowicz-McKinnon K, 18 Martel-Pelletier J, 595 Martin JA, 634 Martini A, 566 Masuda I, 279 Matteson EL, 177, 206 McCarthy GM, 273 Mease PJ, 366 Merkel PA, 31 Michet CJ, Jr, 56 Minor MA, 130 Miossec P, 218 Moldovan I, 499 Müller–Ladner U, 238 Nakamura H, 604 Neumann E, 238 Nirmalananthan N, 684 Nishioka K, 604 Nordborg C, 25 Nordborg E, 25 O’Shea FD, 273 Oatis C, 143 Ollier WER, 707 Østergaard M, 223 Pannu J, 739 Pascual E, 282 Pascual V, 577 Pedraz T, 282 Pelletier J-P, 595 Pilkington C, 673 Plotz PH, 700 Postlethwaite AE, 733 Prahalad S, 588 Rao JK, 119 Reboul P, 595 Reveille JD, 723 Ritchlin CT, 338 Rosandich PA, 192 Rosenquist R, 254 Rosenthal AK, 262 Ruperto N, 566 Ryan LM, 268 Sanz I, 180, 505 Sarzi-Puttini P, 287 Sawalha AH, 534 Scalzi LV, 571 Schmitt WH, 9 Schneider R, 43 Scott DL, 678 Seibl R, 411 Selmi C, 406 Shigemitsu H, 733 Singer NG, 571 Smiles S, 199 Staud R, 157 © 2004 Lippincott Williams & Wilkins ISSN 0268–4705 Stichweh D, 577 Stone MA, 357 Suarez-Almazor ME, 91 Suhrbier A, 374 Sultan SM, 668 Sweeney SE, 231 Tarkowski A, 527 Taylor WJ, 350 Tervaert JWC, 4 Thomas CF, 186 Ting TV, 560 Trojanowska M, 739 Trysberg E, 527 Tsao BP, 513 Tsokos G, 497 Tsokos GC, 548 Turesson C, 206 van der Woude FJ, 9 van Engelen BGM, 692 van Venrooij WJ, 692 Varga J, 714 Vassallo R, 186 Vlieland TPHV, 153 Wakefield RJ, 435 Walsh DA, 609 Walsh NC, 419 Ward MM, 89, 96 Wasko MCM, 109 Weyand CM, 212 Wiell C, 223 Xiang Y, 604 Yazici H, 38 Yurdakul S, 38 Zeidler H, 380 Cumulative Index to Subjects Volume 16 2004 AIMS. See Arthritis Impact Measurement Scales Alphaviruses, 374–379 chikungunya virus, 374 macrophage, 374 Ross River virus, 374 Sindbis virus, 374 viral arthritis, 374 Amyloidoses, 67–75 Amyloid A, 68, 70 Apolipoprotein A1 amyloidosis, 72 Apolipoprotein A2 amyloidosis, 71 immunoglobulin light chain, 69 serum amyloid A, 68, 70 tissue damage, 69 transthyretin amyloid, 70 b2 microglobulin amyloid, 71 treatment, 72 ANCA. See Antineutrophil cytoplasmic antibodies Angiogenesis, 609–615 back pain, 609 hypoxia, 612 innervation, 610 neovascularization, 609 osteoarthritis, 610 spondylosis, 611 Ankylosing Spondylitis Quality of Life Health-related quality of life, 96 Antineutrophil cytoplasmic antibodies, 4–8 animal models, 4–8 mice, 5 rats, 5 antineutrophil cytoplasmic antibody associated vasculitides, 12, 13 B cells, 181, 182 clinical applications, 9–17 assays, 10 enzyme-linked immunoabsorbent assay, 10, 11, 15 target antigens, 10 diagnostic significance, 12 microscopic polyangiitis, 9 pathophysiology, 4–8 relapse rate, 14 specificity, 14 transfer studies, 6 Wegener granulomatosis, 9 Antineutrophil cytoplasmic antibody associated vasculitides antineutrophil cytoplasmic antibodies, 12, 13 AR. See Androgen receptor © 2004 Lippincott Williams & Wilkins ISSN 1040–8711 Arthritis Chlamydia-induced, 380–392 Chlamydia-host cell interaction, 386 Chlamydia molecular diagnostics, 384 HLA-B27, 381 hemochromatosis, 62–66 iron storage disease, 62–66 arthropathy, 62 genetics, 62 HFE genes, 63 pattern recognition receptors, 411–418 caspase recruitment domains, 411, 413 toll-like receptors, 411 triggering receptors expressed by myeloid cells, 411, 414 Arthritis Impact Measurement Scales health-related quality of life, 96 ASQoL. See Ankylosing Spondylitis Quality of Life Azathioprine relapsing polychondritis, 59 sarcoidosis, 54 B cells therapeutic targets, 180–185 antineutrophil cytoplasmic antibodies, 181–183 ANCA-associated vasculitis, 182 cryoglobulinemia, 182 dermatomyositis, 182 rheumatoid arthritis, 181 systemic lupus erythematous, 181 costimulatory molecules, 183 cytokine inhibition, 183 rheumatoid arthritis, 180 Sjögren syndrome, 180 systemic lupus erythematous, 180 systemic sclerosis, 180 Becker muscular dystrophy differential diagnosis, 686, 687 Behcet syndrome antibodies, 39 coagulation abnormalities, 40 infections, 40 autoantibodies, 39 central nervous system, 39 geographical differences, 38 pathogenesis, 39 prognosis, 39 renal involvement, 39 target organ associations, 38 treatment ␣-enolase, 38 anti-TNF, 40, 41 dapsone, 41 IFN-␣, 40 rebamipide, 41 Biologic agents infectious complications, 393–398 tumor necrosis factor, 393 aspergillus species, 395 cryptococcosis, 396 histoplasmosis, 395 listeria monocytogenes, 396 tuberculosis, 394 BMD. See Becker muscular dystrophy Bone biomarkers, 428–434 ankylosing spondylarthritis, 428 biochemical markers, 429 cartilage, 428 inflammatory arthritis, 429 rheumatoid arthritis, 428 Bone mass genetic determinants, 450–456 BS. See Behcet syndrome Calcium crystal deposition diseases, 279–281 calcium pyrophosphate dihydrate, 279 enzymes, 280 hypertrophic chondrocytes, 279 pericellular matrix, 280 transglutaminase, 280 Calcium crystal formation articular cartilage, 263 calcium pyrophosphate dihydrate, 263 crystal models, 264 in vitro models, 263–267 CAM. See Cellular adhesion molecules Cellular adhesion molecules vascular inflammation, 18 Central nervous system vasculitis pediatric, 43–50 angiography, 45 brain biopsy, 46 causes, 44 cerebrospinal fluid, 44 clinical features, 44 epidemiology, 43 imaging, 44, 46 lab tests, 44 post-varicella-zoster virus central nervous system vasculitis, 47 therapy, 46 Cerebrospinal fluid central nervous system, 44 CHAQ. See Childhood Health Assessment Questionnaire Childhood Health Assessment Questionnaire juvenile idiopathic inflammatory myopathies, 674, 675 pediatrics, 102 Childhood Myositis Assessment Scale juvenile idiopathic inflammatory myopathies, 675 CMAS. See Childhood Myositis Assessment Scale CNS. See Central nervous system Computed tomography central nervous system, 44 diffuse idiopathic skeletal hyperostosis, 291 muscle disease, 678, 679 Takayasu arteritis, 34, 35 C-reactive protein vascular inflammation, 21 CRP. See C-reactive protein Crystal arthropathies (overview), 262 CSF. See Cerebrospinal fluid CT. See Computed tomography Dermatomyositis B cells, 182 differential diagnosis, 684 idiopathic inflammatory myopathy, 707 Diffuse idiopathic skeletal hyperostosis, 287–292 computed tomography, 291 etiopathogenesis, 288 magnetic resonance imaging, 291 ossification of ligamentum flavum, 287 ossification of posterior longitudinal ligament, 287 peripheral joint, 288 Disease-modifying antirheumatic drugs rheumatoid arthritis, 193, 207 leflunomide, 194 methotrexate, 193 DM. See Dermatomyositis DMARDs. See Disease-modifying antirheumatic drugs DMD. See Duchenne muscular dystrophy Duchenne muscular dystrophy differential diagnosis, 686 EBV. See Epstein-Barr virus ELISA. See Enzyme-linked immunoabsorbent assay EMS. See Extraskeletal myxoid chondrosarcoma Enzyme-linked immunoabsorbent assay antineutrophil cytoplasmic antibodies, 10, 11, 15 Epstein-Barr virus, 399–405 Hodgkin, 401, 403 immunosuppression, 404 lymphoma, 400 nasopharyngeal carcinoma, 231 non-Hodgkin, 401, 403 rheumatoid arthritis, 399 Evidence-based rheumatology rehabilitation, 130, 131 Fibroblasts functional genomics, 238–245 adhesion, 240 angiogenesis, 240 apoptosis, 242 inflammation, 238 intracellular signaling, 241 matrix degradation, 240 synovial fibroblasts, 238 scleroderma, 733 systemic sclerosis organ fibrosis, 733–738 fibroblast progenitors, 735 TGF-B, 733 tissue-specific fibroblast precursor cells, 734 transdifferentiation, 733 transition, 733 Fibromyalgia. See also Fibromyalgia syndrome exercise, 138–142 fatigue/sleep, 139 intensity, 140 long-term benefits, 141 mood, 139 pain, 139 self-efficacy, 139 setting, 140 types aerobic, 139 anaerobic, 140 flexibility, 140 pain cytokines, 160 fibromyalgia syndrome, 157–159 Hepatitis C virus, 161 HIV infection, 161 infections, 161 neuroendocrine abnormalities, 160 polymyalgia rheumatica, 160 regional musculoskeletal pain, 159. See also myofascial pain Fibromyalgia syndrome genetics, 158 nociception, 158 posttraumatic stress disorder, 158 stressors, 158 triggering events, 158 whiplash, 159 Fibrosing syndromes, 723–732 fibrosing disorders, 728 eosinophilia myalgia syndrome, 728 graft versus host disease, 728 localized scleroderma, 728 nephrogenic fibrosing dermopathy, 728 FMS. See Fibromyalgia syndrome GCA. See Giant cell arteritis Giant cell arteritis, 25–30 clinical manifestations audiovestibular, 25 lower extremity, 26 imaging duplex ultrasonography, 26 FDG-PET, 26 osteoporosis, 28 steroid-sparing agents, 28 corticosteroids, 28 methotrexate, 28 steroid therapy, 28 temporal arteritis, 25, 26 treatment strategies, 26 vasculitis, 2 GJD. See Greutzfeldt-Jakob disease Gout, 282–286 epidemiology, 283 macrophage maturation, 283 treatment, 284 Greutzfeldt-Jakob disease inclusion body myositis, 704 HAQ. See Health Assessment Questionnaire Health Assessment Questionnaire health-related quality of life, 96 Health-related quality of life, 96–101 Ankylosing Spondylitis Quality of Life, 96 Arthritis Impact Measurement Scales, 96 Health Assessment Questionnaire, 96 health status, 96–101 ankylosing spondylitis, 97, 98 osteoarthritis, 96, 98 rheumatoid arthritis, 97, 98 idiopathic inflammatory myopathy, 671 Western Ontario and McMaster Universities Osteoarthritis Index, 96 HLA. See Human leukocyte antigen Host-infectious agent interactions, 371–373 HRQOL. See Health-related quality of life Human leukocyte antigen relapsing polychondritis, 58 IBM. See Inclusion body myositis ICAM-1. See Intracellular adhesion molecule-1 Idiopathic inflammatory myopathy animal models, 711 assessing health-related quality of life, 671 IMACS, 670, 671 visual analogue scale, 670 autoantibodies, 708 cytokine genes, 708 disease mechanisms, 707–713 HLA-DRB1 studies, 707–713 dermatomyositis, 707 HLA-related differences, 708 polymyositis, 707 idiopathic inflammatory myopathy, 710 inclusion body myositis, 710 manual muscle strength testing, 668 microchimerism, 709 muscle biopsy, 669 muscle enzymes, 669 serum creatine kinase, 669 muscle imaging magnetic resonance imaging, 669 magnetic resonance spectroscopy, 669 nonclassical HLA, 708 T cell receptors, 711 IIM. See Idiopathic inflammatory myopathy ILD. See Interstitial lung disease Inclusion body myositis autoimmunity, 703 differential diagnosis, 684, 689 distal myopathy with rimmed vacuoles, 704 endoplasmic reticulum, 704 GNE mutation, 704 Greutzfeldt-Jakob disease, 704 protein folding, 704 unfolded protein response, 704 Inflammatory arthritis, 435–442 bone loss, 419–427 inflammation, 420 IL-1, 422 TNF-␣, 420 osteoclasts, 419 RANKL/RANK/OPG pathway, 422 rheumatoid arthritis, 419 skeletal changes computed tomography, 438 conventional radiography, 435 digital radiogrametry, 441 dual x-ray absorptiometry, 439 ultrasound, 436 Inflammatory biomarkers, 18–24 Interleukin vascular inflammation, 20 Interstitial lung disease rheumatic, 186–191 connective tissue disease, 189 nonspecific interstitial pneumonia, 186 pulmonary hypertension, 188 rheumatoid arthritis, 188 scleroderma, 189 usual interstitial pneumonia, 186 Intracellular adhesion molecule-1 vascular inflammation, 18 JDM. See Juvenile dermatomyositis JIA. See Juvenile idiopathic arthritis JRA. See Juvenile rheumatoid arthritis JSLE. See Juvenile systemic lupus erythematosus Juvenile dermatomyositis pediatrics, 105 Juvenile idiopathic arthritis cardiovascular disease, 110 genetics, 588–594 pediatrics, 102 Juvenile idiopathic inflammatory myopathies Childhood Health Assessment Questionnaire, 674, 675 Childhood Myositis Assessment Scale, 675 Disease Activity Score, 675 juvenile dermatomyositis, 673 incidence, 674 pediatric assessment tools, 674 pulmonary, 674 magnetic resonance imaging scan, 674 Juvenile rheumatoid arthritis Childhood Health Assessment Questionnaire, 102 disability, 104 osteopenia, 105 pediatrics, 103 remission, 103 Juvenile systemic lupus erythematosus, 106 Knee osteoarthritis physical therapy, 143–147 manual therapy, 143 transcutaneous electrical nerve stimulation, 144 LGMD. See Limb-girdle muscular dystrophy Limb-girdle muscular dystrophy differential diagnosis, 686, 687 Lipid metabolism, 76–79 musculoskeletal features, 76–79 fenofibrate, 77 primary hyperlipidemia, 77 Magnetic resonance imaging central nervous system, 44 diffuse idiopathic skeletal hyperostosis, 291 idiopathic inflammatory myopathy, 669 muscle disease, 678, 679 Takayasu arteritis, 31–33 Magnetic resonance spectroscopy idiopathic inflammatory myopathy, 669 muscle disease, 678–680 Malignant lymphomas, 254–261 anti-TNF, 258 DMARDs, 257–259 TNF-blocking therapy, 254 MCP. See Monocyte chemoattractant protein-1 MCH. See Multicentric reticulohistiocytosis Methotrexate giant cell arteritis, 28 relapsing polychondritis, 59 sarcoidosis, 54 Microscopic polyangiitis antineutrophil cytoplasmic antibodies, 9 Modes of practice case mix, 125 patient-centered care, 125 primary care, 125 Monocyte chemoattractant protein-1 vascular inflammation, 19 MPA. See Microscopic polyangiitis mPGES-1, 623–627 prostaglandin E2, 623 MRI. See Magnetic resonance imaging MSA. See Myositis specific autoantibodies MTX. See Methotrexate Multicentric reticulohistiocytosis, 76–79 musculoskeletal features, 76–79 fenofibrate, 77 primary hyperlipidemia, 77 Muscle disease idiopathic inflammatory myopathies, 679 imaging, 678, 679 computed tomography, 678,679 magnetic resonance imaging, 678, 679 magnetic resonance spectroscopy, 678–680 scintigraphy, 679 ultrasound, 679 x-rays, 679 calcinosis, 679 myositis ossificans, 769 muscle infarction, 680 myositis, 680 polymyositis, 679 range muscle infarction, 678 myositis ossificans, 678 Musculoskeletal aging, 114–118 osteoarthritis, 114 osteoporosis, 115 hip fracture, 115 treatment, 115 sarcopenia, 116 self-management, 117 Myositis cellular immune mechanisms, 701 clinical features, 685 co-stimulatory molecules, 702 differential diagnosis, 684–691 Becker muscular dystrophy, 686, 687 dermatomyositis, 684, 700 Duchenne muscular dystrophy, 686 dystrophinopathy, 687 endocrine myopathies, 689 inclusion body myositis, 684, 689, 700 Limb-girdle muscular dystrophy, 686, 687 lipid storage disorders, 689 metabolic myopathies, 688 mitochondrial myopathies, 689 muscle glycogenoses, 688 disease assessment, 665 electromyography, 685 European League against Rheumatism, 666 International Myositis Assessment and Clinical Studies, 665 interstitial lung disease, 701 muscle biopsy, 686 myosin heavy chain, 686, 702 myositis damage index, 666 myositis damage score, 666 necrotizing myopathy, 701 polymyositis, 684, 700 transglutaminase 2, 703 Myositis specific autoantibodies, 692–699 anti-signal recognition particle, 696 diagnosis criteria, 692 immunopathogenesis, 696 Jo-1, 694–696 myositis associated autoantibodies, 692 Nonsteroidal antiinflammatory drug relapsing polychondritis, 59 sarcoidosis, 54 NSAID. See Nonsteroidal antiinflammatory drug Osteoarthritis. Also see specific locations calcium phosphate deposition, 273–278 basic calcium phosphate crystals, 273 intracellular calcium signaling, 275 matrix metalloproteinase, 273, 275, 277 mitogenesis, 274 phosphocitrate, 276 tissue inhibitors of metalloproteinases, 275 cartilage, 604–608 neoantigens, 604 galactin-3, 595–598 mesenchymal stem cells, 599–603 musculoskeletal aging, 114 pain, 628–633 acetaminophen, 631 cyclooxygenase-2, 629 lipoxygenase, 632 NSAIDs, 630 signaling transduction, 616–622 sports, 634–637 articular cartilage, 634 posttraumatic osteoarthritis, 634 Osteoclasts, 464–468 B cell, 465 lineage development, 464 transcriptional regulation, 465 Osteonecrosis, 443–449 avascular necrosis, 443 diseases of the hip, 443 Osteoporosis musculoskeletal aging, 115 Parathyroid hormone, 457–463 hormone replacement therapy, 460 osteoanabolic agent, 457 osteoporosis, 459 Patient-centered care, 89, 90 Patient-physician communication, 91–95 health services research, 91 medical eduction, 91 rheumatic disease, 91 PCNSL. See Primary central nervous system lymphoma Pediatric rheumatology Health Assessment Questionnaire, 102 juvenile dermatomyositis, 105 juvenile idiopathic arthritis, 102 juvenile rheumatoid arthritis, 103 juvenile systemic lupus erythematosus, 106 psychosocial aspects, 555–559 remittive agents, 571–576 DMARDs, 571 methotrexate, 571 mycophenolate mofetil, 572 Pediatric Rheumatology International Trials Organisation, 565–570, 676 Childhood Health Assessment Questionnaire, 674, 675 Childhood Myositis Assessment Scale, 675 PET. See Positron emission tomography PH. See Pulmonary hypertension PKC. See Protein kinase C PM. See Polymyositis PMR. See Polymyalgia rheumatica Polymyalgia rheumatica fibromyalgia pain, 160 Polymyositis differential diagnosis, 684 idiopathic inflammatory myopathy, 707 Positron emission tomography central nervous system, 46 Takayasu arteritis, 36 Primary biliary cirrhosis, 406–410 autoimmunity, 407 Guillain-Barre syndrome, 407 HLA-B27, 407 Spondyloarthropathies, 407 bacteria, 408 Escherichia coli, 408 Helicobacter species, 409 Sphingomonas species, 408 Primary central nervous system lymphoma, 453, 601–606 delayed neurotoxicity, 603 epidemiology, 601 pathogenesis, 601 prognostic factors, 602 rituximab, 601, 604 treatment, 602 elderly, 603 Primary hyperparathyroidism, 1–7 etiology, 1 genetics, 2 histopathology, 2 parathyroid gland, 1 surgery, 3 Protein kinase C glucocorticoid-induced apoptosis, 556 scleroderma, 743 Psoriasis vulgaris dendritic cells, 331 genomics, 333 inflammatory cytokines, 331 leukocytes, 331 natural killer T cells, 334 susceptibility genes, 335 T lymphocytes, 331 Psoriatic arthritis, 338–343 abnormal bone remodeling, 341 acquired immune response, 340 angiogenesis, 341 genetic factors, 338 human leukocyte antigen, 338, 339 innate immune response, 340 leflunomide, 362 methotrexate, 361 outcome assessment, 350–356 core domains, 351 measurement instrument, 351 OMERACT, 350 quality of life, 354 responder criteria, 355 pathogenesis, 338 spondyloarthropathies, 344–349 sulphasalazine, 362 TNF blockers, 362 Pulmonary hypertension rheumatic interstitial lung disease, 188 RA. See Rheumatoid arthritis Radiation Therapy and Oncology Group head and neck squamous cell carcinoma, 217 metastatic lesions, 309 prostate cancer, 243 Raynaud phenomenon fibrosing disorders, 723 scleroderma, 718–722 Relapsing polychondritis, 56–61 clinical features cardiovascular, 58 dermatologic, 57 musculoskeletal, 57 neurologic, 58 otorhinolaryngeal, 57 pulmonary, 57 renal, 57 differential diagnosis, 59 disease associations, 58 epidemiology, 58 management azathioprine, 59 biologic, 59 methotrexate, 59 NSAID, 59 pathogenesis HLA-DR4, 58 Rheumatic diseases comorbid conditions, 109–112 cardiovascular disease, 110 juvenile idiopathic arthritis, 110 infection, 109 malignancy, 112 prevention, 119–124 behavioral, 120 cognitive, 120 exercise, 121 obesity, 121 occupational injury, 122 Rheumatoid arthritis biologic response modifiers, 192–198 IL-1 receptor antagonist, 196 medications DMARDs, 193 glucocorticoids, 193 NSAIDs, 193 TNF-␣, 194 comorbidity, 177 managing, 177–179 NSAIDs, 177 cytokine network, 218–222 employment, 148–152 extra-articular disease manifestations cardiac, 206 hematologic, 207 nonpharmacologic treatments, 209 pulmonary, 206 TNF inhibitors, 208 vasculitis, 208 long-term risks, 199–205 demyelination, 203 heart failure, 203 infections, 199 bacterial, 202 myobacterium tuberculosis, 201 opportunistic, 202 lymphoma, 202 systemic lupus erythematosus-like syndromes, 203 TNF-␣ antagonists adalimumab, 199 etanercept, 199 infliximab, 199 multidisciplinary team care, 153–156 clinical nurse specialist, 154 inpatient vs. day patient, 153 interdisciplinary communication, 155 outcome assessment, 155 signal transduction, 231–237 mitogen-activated protein kinase, 231 c-Jun-N-terminal kinase, 232 extracellular signal-related kinases, 232 p38, 231 NF-, 232 Janus kinase, 233 signal transducer activator of transcription, 233 toll-like receptors, 234 strength training, 132–137 muscle function, 132 principles, 132 safety, 136 scientific evidence, 135 T-cell regulation, 212–217 regulatory T cells, 213 autoantibodies, 214 CD-28 mediated costimulation, 213 novel costimulatory pathways, 214 senescent CD4 T cells, 214 thymic function, 212 ultrasonography, 223–230 work disability, 148–152 assessing, 149 Work Instability Scale, 149 Work Limitations Questionnaire, 149, 150 prevalence, 148 return-to-work, 150 risk factors, 149 work retention, 150 Rheumatoids factor, 246–253 autoimmunity, 246 germinal centers, 246 B cell, 250 SAA. See Serum amyloid A, 68, 70 Sarcoidosis rheumatologic manifestations, 51–55 general clinical features diagnosis, 52 granulomas, 51 rheumatology, 52 acute arthritis, 53 bone, 53 chronic arthritis, 53 myopathy, 53 treatment antimalarials, 54 azathioprine, 54 cyclosporine, 54 methotrexate, 54 NSAIDs, 54 TNF, 54 Scleroderma connective tissue disease, 718 endothelial-dependent relaxation, 719 genetic factors, 719 fibroblasts, 733 signaling, 739–745 fibrosis, 742 protein kinase C pathway, 743 rapamycin pathway, 743 Smad 2/3 pathway, 742 TGF-B signaling, 739 fibrinolysis, 719 angiogenesis, 720 atherosclerosis, 719 circulating endothelial cells, 720 endothelial apoptosis, 720 gene expression profile, 720 hyperlipidemia, 719 vasculogenesis, 720 immune involvement cytokines, 720 lymphocyte transendothelial migration, 720 Raynaud phenomenon, 718–722 vascular disease, 718–722 Serum amyloid A amyloidoses, 68, 70 Spondyloarthropathy autoimmunity, 407 treatment, 357–365 ankylosing spondylitis, 357 DMARD, 358 exercise, 358 NSAIDs, 358 pamidronate, 358 thalidomide, 359 TNF blockers, 359 psoriatic arthritis leflunomide, 362 methotrexate, 361 sulphasalazine, 362 TNF blockers, 362 STAT. See Signal transducer activator of transcription Signal transducer activator of transcription rheumatoid arthritis, 233 Sjögren syndrome apoptosis, 522–526 B cells, 180 estrogen deficiency, 522, 523 T-cell apoptosis, 523 rheumatic interstitial lung disease, 187, 189 systemic lupus erythematosus, 497, 498 SLE. See Systemic lupus erythematosus Spondyloarthropathies (overview), 329, 330 STAT. See Signal transducer activator of transcription Systemic lupus erythematosus antibodies, 181 antinuclear antibodies, 534–540 B cells, 180 human, 505 murine, 505 cerebral inflammation, 527–533 biochemistry, 528 central nervous system, 527 cytokines, 528 metalloproteinases, 527 neuroimaging, 530 genetics, 513–521 association, 513 gene variants, 515–519 linkage, 513 Interferon-␣, 541–547 overview, 497, 498 pediatric, 577–587 T lymphocytes, 548–552 chemokine receptors, 549 co-stimulatory molecules, 459 intracellular signaling, 550 T-cell receptor, 548 trials biologics, 501 immunoablation, 502 immunosuppressive therapy, 500 nonpharmacologic, 503 SELENA, 499 Systemic sclerosis, 723–732 animal models, 746–752 bleomycin-induced scleroderma, 746 pathogenesis, 746 Tsk mice, 746 pulmonary fibrosis, 749 TGF-B, 746 transgenic mice, 750 autoantibodies antibodies against extractable nuclear antigens, 727 anticentromere antibodies, 724 antinuclear antibodies, 725 antiphospholipid antibodies, 727 antitopoisomerase 1 antibodies, 725 TAK. See Takayasu arteritis Takayasu arteritis diagnostic imaging, 31–37 computed tomography scan, 34 diagnosis, 34 disease activity, 35 limitations, 35 Doppler ultrasound, 33 diagnosis, 33 disease activity, 34 limitations, 34 magnetic resonance imaging/angiography, 31 diagnosis, 31 disease activity, 34 limitations, 34 positron emission tomography diagnosis, 36 disease activity, 36 limitations, 36 TLR. See Toll-like receptors Toll-like receptors rheumatoid arthritis, 234 Transcranial Doppler ultrasound central nervous system, 46 Transthyretin amyloid amyloidoses, 70 TSC. See Tuberous sclerosis complex TTR. See Transthyretin amyloid Tuberous sclerosis complex renal cell carcinoma, 247 Varicella-Zoster virus central nervous system, 47 Vascular cellular adhesion molecule-1 vascular inflammation, 18 Vascular inflammation, 18–24 accessory signaling molecules acute phase proteins, 21 C-reactive protein, 21 cellular adhesion molecules, 18 chemokines, 19 monocyte chemoattractant protein-1, 19 cytokines, 19 Interleukins, 20 TNF-␣, 19 intercellular adhesion molecule-1, 18 interleukin, 20 IL-1, 20 IL-6, 20 IL-10, 20 IL-18, 20 proteases matrix metalloproteinases, 21 vascular cellular adhesion molecule-1, 18 Vasculitides childhood, 560–565 Behcet disease, 563 Central nervous system vasculitis, 563 Henoch-Schönlein purpura, 560 Secondary vasculitis, 563 Takayasu arteritis, 562 Wegener granulomatosis, 562 Vasculitis, 1 anti-neutrophil cytoplasmic antibodies, 1 Behcet syndrome, 2 central nervous system, 2 giant cell arteritis, 2 inflammatory biomarkers, 3 C-reactive protein, 3 Takayasu arteritis, 2 VCAM-1. See Vascular cellular adhesion molecule-1 VZV. See Varicella-Zoster virus Wegener granulomatosis antineutrophil cytoplasmic antibodies, 9 WG. See Wegener granulomatosis Western Ontario and McMaster Universities Osteoarthritis Index health-related quality of life, 96 Current Opinion in RHEUMATOLOGY Reviews of all advances ⴢ Evaluations of key references Comprehensive listing of papers Volume 16 ⴢ 2004 Lippincott Williams & Wilkins Copyright © 2004 Lippincott Williams & Wilkins ISSN 1040–8711 Current Opinion in RHEUMATOLOGY Volume 16 Number 1 January 2004 The systemic amyloidoses Joel N Buxbaum 67 Vasculitis syndromes Musculoskeletal features of disorders of lipid metabolism and multicentric reticulohistiocytosis Thomas Bardin 76 Edited by Carol A Langford Editorial overview: vasculitis: from pathophysiology to clinical applications Carol A Langford 1 Antineutrophil cytoplasmic autoantibodies and pathophysiology: new insights from animal models Dennis Huugen, Jan Willem Cohen Tervaert and Peter Heeringa 4 Clinical applications of antineutrophil cytoplasmic antibody testing Wilhelm H Schmitt and Fokko J van der Woude 9 Current world literature Vasculitis syndromes 80 Systemic disorders with rheumatic manifestations 84 List of journals scanned Recent advances in vascular inflammation: C-reactive protein and other inflammatory biomarkers Kathleen Maksimowicz-McKinnon, Deepak L Bhatt and Leonard H Calabrese 18 Giant cell arteritis: strategies in diagnosis and treatment Elisabeth Nordborg and Claes Nordborg 25 Diagnostic imaging in Takayasu arteritis Eugene Y Kissin and Peter A Merkel Number 2 March 2004 Epidemiology and health-related services Edited by Michael Ward Patient-centered care and health outcomes Michael M Ward 89 Patient-physician communication Maria E Suarez-Almazor 91 96 31 Outcome measurement: health status and quality of life Michael M Ward Behçet syndrome Sebahattin Yurdakul, Vedat Hamuryudan and Hasan Yazici 38 Health outcomes in pediatric rheumatic diseases Ciaran M Duffy 102 109 Central nervous system vasculitis in children Susanne Benseler and Rayfel Schneider 43 Comorbid conditions in patients with rheumatic diseases: an update Mary Chester M Wasko Musculoskeletal aging Suzanne G Leveille 114 Prevention research and rheumatic disease Jaya K Rao and Jennifer M Hootman 119 125 Systemic disorders with rheumatic manifestations Edited by Doyt Ladean Conn Rheumatologic manifestations of sarcoidosis Andy Abril and Marc D Cohen 51 New modes of practice John R Kirwan Relapsing polychondritis Peter D Kent, Clement J Michet, Jr and Harvinder S Luthra 56 Rehabilitation medicine in rheumatic diseases Arthritis in hemochromatosis or iron storage disease Joanne M Jordan 62 Edited by Marian A Minor Editorial overview: meeting the challenges of evidence-based rheumatology rehabilitation Marian A Minor 130 Effectiveness and safety of strength training in rheumatoid arthritis Arja Häkkinen 132 Effectiveness of exercise in management of fibromyalgia Susan E Gowans and Amy deHueck 138 Role of physical therapy in management of knee osteoarthritis G Kelley Fitzgerald and Carol Oatis 143 Employment and work disability in rheumatoid arthritis Catherine L Backman Rheumatoid arthritis Edited by Gerd R Burmester T-cell regulation in rheumatoid arthritis Jörg J Goronzy and Cornelia M Weyand 212 An update on the cytokine network in rheumatoid arthritis Pierre Miossec 218 223 148 Ultrasonography in rheumatoid arthritis: a very promising method still needing more validation Mikkel Østergaard and Charlotte Wiell 153 Signal transduction in rheumatoid arthritis Susan E Sweeney and Gary S Firestein 231 Multidisciplinary team care and outcomes in rheumatoid arthritis Thea PM Vliet Vlieland 238 Fibromyalgia pain: do we know the source? Roland Staud 157 Functional genomics of fibroblasts Elena Neumann, Renate E Gay, Steffen Gay and Ulf Müller–Ladner Rheumatoid factor revisited Thomas Dörner, Karl Egerer, Eugen Feist and Gerd R Burmester 246 Rheumatoid arthritis and malignant lymphomas Eva Baecklund, Johan Askling, Richard Rosenquist, Anders Ekbom and Lars Klareskog 254 Current world literature Epidemiology and health-related services 164 Rehabilitation medicine in rheumatic diseases 171 List of journals scanned Crystal deposition diseases Edited by Ann K Rosenthal Number 3 May 2004 Clinical therapeutics Editorial overview: crystal arthropathies and other unpopular rheumatic diseases Ann K Rosenthal 262 In vitro models of calcium crystal formation Claudia Gohr 263 268 Editorial overview: managing comorbidity risks in rheumatoid arthritis Sonja A Dechant and Eric L Matteson 177 Modulation of chondrocyte production of extracellular inorganic pyrophosphate Jill C Costello and Lawrence M Ryan 273 B cells as therapeutic targets for rheumatic diseases R John Looney, Jennifer Anolik and Inãki Sanz 180 Basic calcium phosphate deposition in the joint: a potential therapeutic target in osteoarthritis Finbar D O’Shea and Geraldine M McCarthy 279 Advances in the treatment of rheumatic interstitial lung disease Robert Vassallo and Charles F Thomas 186 Calcium crystal deposition diseases: lessons from histochemistry Ikuko Masuda Gout Eliseo Pascual and Teresa Pedraz 282 Perioperative management of patients with rheumatoid arthritis in the era of biologic response modifiers Peter A Rosandich, Joe T Kelley, III and Doyt L Conn 192 New developments in our understanding of DISH (diffuse idiopathic skeletal hyperostosis) Piercarlo Sarzi-Puttini and Fabiola Atzeni 287 Long-term risks associated with biologic response modifiers used in rheumatic diseases Anna K Imperato, Stephen Smiles and Steven B Abramson 199 Management of extra-articular disease manifestations in rheumatoid arthritis Carl Turesson and Eric L Matteson 206 Edited by Eric L Matteson Current world literature Clinical therapeutics 293 Rheumatoid arthritis 305 Crystal deposition diseases 324 List of journals scanned Metabolic bone disease Number 4 July 2004 Edited by Steven Goldring Spondyloarthropathies Edited by Dafna D Gladman Editorial overview: spondyloarthropathies Dafna D Gladman 329 Psoriasis vulgaris: an interplay of T lymphocytes, dendritic cells, and inflammatory cytokines in pathogenesis Francesca Chamian and James G Krueger 331 Pathogenesis of psoriatic arthritis Allen P Anandarajah and Christopher T Ritchlin 338 Relationship of psoriatic arthritis with the other spondyloarthropathies Philip S Helliwell 344 Assessment of outcome in psoriatic arthritis William J Taylor 350 Recent advances in the treatment of the spondyloarthropathies Yan Liu, Daniela Cortinovis and Millicent A Stone 357 Recent advances in the management of psoriatic arthritis Philip J Mease 366 Bone loss in inflammatory arthritis: mechanisms and treatment strategies Nicole C Walsh and Ellen M Gravallese 419 Noninvasive techniques for assessing skeletal changes in inflammatory arthritis: bone biomarkers Patrick Garnero and Pierre D Delmas 428 Noninvasive techniques for assessing skeletal changes in inflammatory arthritis: imaging technique Richard J Wakefield, Philip G Conaghan, Steve Jarrett and Paul Emery 435 Osteonecrosis: etiology, diagnosis, and treatment Lynne C Jones and David S Hungerford 443 Genetic determinants of bone mass PA Baldock and John A Eisman 450 Parathyroid hormone: evolving therapeutic concepts Nancy E Lane 457 The origins of osteoclasts Mark C Horowitz and Joseph A Lorenzo 464 Current world literature Spondyloarthropathies 469 Infectious arthritis and immune dysfunction Infectious arthritis and immune dysfunction 475 Edited by JS Hill Gaston Metabolic bone disease 482 Editorial overview: host–infectious agent interactions in the pathogenesis of rheumatic disease JS Hill Gaston 371 Clinical and pathologic aspects of arthritis due to Ross River virus and other alphaviruses Andreas Suhrbier and May La Linn 374 Chlamydia-induced arthritis Henning Zeidler, Jens Kuipers and Lars Köhler 380 Infectious complications of treatment with biologic agents Carol Dukes Hamilton 393 Epstein-Barr virus, arthritis, and the development of lymphoma in arthritis patients Margaret FC Callan List of journals scanned Number 5 September 2004 Systemic lupus erythematosus and Sjögren syndrome Edited by George Tsokos Editorial overview: SLE and Sjögren syndrome in 2004 George Tsokos 497 499 399 Systemic lupus erythematosus trials: successes and issues Ellen M Ginzler and Ioana Moldovan 505 Bacteria and human autoimmunity: the case of primary biliary cirrhosis Carlo Selmi and M Eric Gershwin 406 B cells in human and murine systemic lupus erythematosus Jennifer Anolik and Iñaki Sanz 513 Pattern recognition receptors and their involvement in the pathogenesis of arthritis Reinhart Seibl, Diego Kyburz, Roger P Lauener and Steffen Gay 411 Update on human systemic lupus erythematosus genetics Betty P Tsao Apoptosis and estrogen deficiency in primary Sjögren syndrome Yoshio Hayashi, Rieko Arakaki and Naozumi Ishimaru 522 mPGES-628s a novel target for arthritis Hassan Fahmi 623 Pain and osteoarthritis: new drugs and mechanisms Burkhard Hinz and Kay Brune 628 Sports and osteoarthritis Joseph A Buckwalter and James A Martin 634 541 548 Current world literature Cerebral inflammation and degeneration in systemic lupus erythematosus Estelle Trysberg and Andrej Tarkowski 527 Antinuclear autoantibodies in systemic lupus erythematosus Amr H Sawalha and John B Harley 534 Interferon-␣ in systemic lupus erythematosus Mary K Crow and Kyriakos A Kirou T lymphocytes in systemic lupus erythematosus: an update Vasileios C Kyttaris and George C Tsokos Systemic lupus erythematosus and Sjögren syndrome 640 Pediatric and heritable disorders Pediatric and heritable disorders 649 Edited by Raphael Hirsch Osteoarthritis 652 Editorial overview: pediatric rheumatology workforce: a status update Raphael Hirsch 553 List of journals scanned Psychosocial aspects in pediatric rheumatology Daniel Kietz 555 Number 6 November 2004 Update on childhood vasculitides Tracy V Ting and Philip J Hashkes 560 Myositis and myopathies International research networks in pediatric rheumatology: the PRINTO perspective Nicolino Ruperto and Alberto Martini 566 Remittive agents in pediatric rheumatology Nora G Singer and Lisabeth V Scalzi 571 Update on pediatric systemic lupus erythematosus Dorothee Stichweh, Edsel Arce and Virginia Pascual 577 Genetics of juvenile idiopathic arthritis: an update Sampath Prahalad 588 Edited by David Isenberg Osteoarthritis Edited by Johanne Martel-Pelletier and Jean-Pierre Pelletier Editorial overview: making sure the treatment of myositis does not get “lost in translation” David Isenberg 665 Clinical assessment in adult onset idiopathic inflammatory myopathy SM Sultan 668 Clinical assessment in juvenile idiopathic inflammatory myopathies and the development of disease activity and damage tools Clarissa Pilkington 673 Use of imaging to assess patients with muscle disease David L Scott and Gabrielle H Kingsley 678 684 Editorial overview: galectin-3 in osteoarthritis: when the fountain of youth doesn’t deliver its promises Pascal Reboul, Johanne Martel-Pelletier and Jean-Pierre Pelletier 595 Is it really myositis? A consideration of the differential diagnosis Niranjanan Nirmalananthan, Janice L Holton and Michael G Hanna 692 Mesenchymal stem cells in osteoarthritis Frank P Luyten 599 Myositis specific autoantibodies: changing insights in pathophysiology and clinical associations Gerald JD Hengstman, Baziel GM van Engelen and Walther J van Venrooij Neoantigens in osteoarthritic cartilage Tomohiro Kato, Y Xiang, H Nakamura and K Nishioka 604 Myositis: an update on pathogenesis Lisa Christopher-Stine and Paul H Plotz 700 Angiogenesis in osteoarthritis and spondylosis: successful repair with undesirable outcomes David A Walsh 609 707 Signaling transduction: target in osteoarthritis Francis Berenbaum 616 Have recent immunogenetic investigations increased our understanding of disease mechanisms in the idiopathic inflammatory myopathies? Hector Chinoy, William ER Ollier and Robert G Cooper Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes Edited by John Varga Editorial overview: illness and art: the legacy of Paul Klee John Varga 714 Raynaud phenomenon and the vascular disease in scleroderma M Bashar Kahaleh 718 Autoantibodies in systemic sclerosis and fibrosing syndromes: clinical indications and relevance Eduardo J Cepeda and John D Reveille 723 Cellular origins of fibroblasts: possible implications for organ fibrosis in systemic sclerosis 733 Arnold E Postlethwaite, Hidenobu Shigemitsu and Siva Kanangat Recent advances in fibroblast signaling and biology in scleroderma Jaspreet Pannu and Maria Trojanowska 739 Animal models of systemic sclerosis: insights into systemic sclerosis pathogenesis and potential therapeutic approaches Paul J Christner and Sergio A Jimenez 746 Erratum 753 Current world literature Myositis and myopathies 754 Raynaud phenomenon, scleroderma, overlap syndromes, and other fibrosing syndromes 767 List of journals scanned I ﺷــﺮﻛـﺖ ﺭﻫــﺮﻭﺍﻥ ﻃــﺐ ((ﺛﻤﻴﻦ ))ﺑﺎ ﻣﺴﺌﻮﻟﻴﺖ ﻣﺤﺪﻭﺩ Medical Journals Print & CD-Rom Full Text Archives of 250 Medical Journals of CD-Rom Printable , Color For More Info. Please Contact Us : Tel & Fax : 021- 6429069 - 6432484 – 6947341 Second Floor , NO.135,Keshavarz Blvd./ Tehran 14187 , Iran – P.O.Box : 14185 - 157